Fact-checked by Grok 2 weeks ago

Tenapanor

Tenapanor is a small-molecule that acts as a selective of the sodium/ exchanger 3 (NHE3) in the , primarily used to treat with (IBS-C) in adults and to lower levels in adults with (CKD) on who have an inadequate response to or intolerance of binders. By blocking NHE3, tenapanor reduces sodium absorption in the intestines, which promotes water retention in the intestinal lumen, softens stool, and accelerates gastrointestinal transit for its effects in IBS-C, while also decreasing paracellular absorption in the CKD indication. Developed by the company Ardelyx, tenapanor was first approved by the U.S. (FDA) on September 12, 2019, under the brand name IBSRELA for the treatment of IBS-C, based on two pivotal phase 3 clinical trials (T3MPO-1 and T3MPO-2) that demonstrated significant improvements in complete spontaneous bowel movements and compared to . On October 17, 2023, the FDA granted approval for its second indication under the brand name XPHOZAH, supported by three phase 3 trials (PHREEDOM, , and AMPLIFY) showing reductions in phosphorus levels of approximately 0.7 mg/dL versus when added to standard therapy. It has also received approvals in (2020, for IBS-C), (2023, for hyperphosphatemia), and (November 2025, for hyperphosphatemia). Marketed in tablet form at strengths of 50 mg for IBSRELA and 10 mg, 20 mg, or 30 mg for XPHOZAH, it is administered orally twice daily immediately before meals, with common side effects including (affecting up to 53% of users, sometimes severe), , , and ; severe may lead to , necessitating dose suspension or discontinuation. Contraindicated in patients under 6 years due to risk of life-threatening observed in and in those with mechanical gastrointestinal obstruction, tenapanor represents a mechanism for managing these conditions without systemic absorption or direct action.

Medical Uses

Irritable Bowel Syndrome with Constipation

Irritable bowel syndrome with constipation (IBS-C) is a subtype of characterized by recurrent abdominal pain at least one day per week in the last three months, associated with two or more of the following: related to , change in frequency of , or change in form (appearance) of , with hard or lumpy more than 25% of the time and loose or watery less than 25% of the time. Patients typically experience fewer than three spontaneous bowel movements per week, often accompanied by straining, incomplete evacuation, , and abdominal discomfort. Tenapanor addresses these symptoms by inhibiting the sodium/hydrogen exchanger 3 (NHE3) in the , which reduces sodium absorption and increases fluid secretion into the bowel, thereby enhancing bowel movement frequency and improving consistency. The recommended dosing for tenapanor in adults with IBS-C is 50 mg taken orally twice daily, immediately prior to the morning and evening meals. This regimen was evaluated in two pivotal phase 3, double-blind, placebo-controlled trials (T3MPO-1 and T3MPO-2) involving over 1,800 patients meeting III criteria for IBS-C, with baseline characteristics including fewer than three complete spontaneous bowel movements (CSBMs) per week and moderate . In T3MPO-1, a 12-week , 27% of tenapanor-treated patients were overall responders (defined as ≥30% reduction in worst and increase of ≥1 CSBM per week for at least 6 of the first 12 weeks) compared to 19% on (P=0.02). In T3MPO-2, a 26-week , 37% of tenapanor-treated patients achieved overall responder status compared to 24% on (P<0.001). Key secondary endpoints showed higher CSBM responder rates (increase of ≥1 CSBM per week for ≥6/12 weeks) of 34% versus 29% in T3MPO-1 and 47% versus 33% in T3MPO-2, alongside improvements in responders (≥30% reduction for ≥6/12 weeks) of 44% versus 33% and 50% versus 38%, respectively. These results demonstrate tenapanor's efficacy in increasing CSBM frequency and reducing pain, with benefits observed as early as week 1 and sustained throughout treatment. Long-term safety was assessed in the T3MPO-3 open-label extension study, where patients who completed T3MPO-1 or T3MPO-2 received tenapanor for up to 52 weeks total exposure. Of 318 enrolled patients, 245 (77%) completed 40 weeks of extension treatment, with 90 receiving ≥52 weeks overall; the safety profile remained consistent with shorter-term studies, primarily featuring diarrhea in 16% (severe in 2.5%), abdominal distension in 3%, flatulence in 3%, and dizziness in 2%, leading to discontinuation in 7.6% (mostly due to diarrhea). No new safety signals emerged, supporting tenapanor's tolerability for chronic use in IBS-C. Patient selection for tenapanor requires confirmation of IBS-C diagnosis via , with exclusion of those with known or suspected mechanical gastrointestinal obstruction, as it is contraindicated in such cases due to risk of bowel perforation or obstruction exacerbation. It is also contraindicated in children under 6 years and not recommended for ages 6 to under 12 due to dehydration risk observed in juvenile animal studies; adults should be monitored for severe diarrhea, with dosing suspension and rehydration if needed.

Hyperphosphatemia in Chronic Kidney Disease

Hyperphosphatemia is a common complication in patients with chronic kidney disease (CKD) on maintenance dialysis, affecting up to 75% of this population due to diminished renal phosphate clearance and enhanced gastrointestinal absorption. Elevated serum phosphorus levels contribute to adverse outcomes, including accelerated vascular calcification, cardiovascular events, secondary hyperparathyroidism, and heightened all-cause mortality risk. Oral phosphate binders represent the primary pharmacologic intervention, forming insoluble complexes with dietary phosphate in the intestine to limit absorption. Despite their widespread use, binders are limited by high daily pill burdens (often 9–12 tablets per meal), frequent gastrointestinal side effects like nausea and constipation, reduced patient adherence, and failure to achieve target phosphorus levels in approximately 50% of patients. Tenapanor is indicated as add-on therapy to phosphate binders for reducing serum phosphorus in adults with CKD on dialysis who exhibit inadequate control or intolerance to binders alone; it may also serve as monotherapy for those unable to tolerate binders. By selectively inhibiting the sodium/hydrogen exchanger 3 (NHE3) in the gut, tenapanor decreases paracellular permeability and thereby reduces dietary phosphate uptake. The recommended dosing regimen is 30 mg orally twice daily, administered immediately prior to the morning and evening meals. In cases of severe diarrhea, the dose may be reduced to 20 mg twice daily to improve tolerability while maintaining efficacy. Efficacy data from pivotal phase 3 trials support tenapanor's role in phosphorus management. In the , a 52-week randomized, placebo-controlled study evaluating tenapanor monotherapy in dialysis patients, tenapanor resulted in a mean serum phosphorus reduction of 1.4 mg/dL versus an increase on placebo. The , conducted in Chinese patients with end-stage renal disease on hemodialysis, demonstrated significant reductions in serum phosphorus with tenapanor compared to placebo. In the , a 4-week randomized, placebo-controlled study of tenapanor as add-on therapy to existing binders, 37% of tenapanor-treated patients achieved serum phosphorus <5.5 mg/dL versus 22% on placebo, with statistically significant mean phosphorus changes favoring tenapanor (-0.72 mg/dL vs. -0.19 mg/dL). A 2024 open-label OPTIMIZE trial further showed that tenapanor as add-on therapy reduced the need for phosphate binders (mean decrease of 3.1 pills/day) while achieving serum phosphorus ≤5.5 mg/dL in 34–38% of patients over 10 weeks. Post-initiation monitoring of serum phosphorus is essential, with levels assessed at baseline, 2–4 weeks after starting therapy, and monthly thereafter, or more frequently if clinically indicated. Dose adjustments should aim to maintain phosphorus within the KDIGO target of 3.5–5.5 mg/dL, balancing efficacy against gastrointestinal tolerability.

Pharmacology

Mechanism of Action

Tenapanor is a selective, non-absorbed small-molecule inhibitor of the sodium/hydrogen exchanger isoform 3 (NHE3), a membrane antiporter predominantly expressed on the apical surface of intestinal epithelial cells in the small intestine and colon. NHE3 facilitates electroneutral sodium absorption by exchanging intracellular protons (H⁺) for extracellular sodium ions (Na⁺) from the intestinal lumen, which drives the coupled absorption of water and electrolytes, contributing to overall fluid homeostasis in the gastrointestinal tract. By specifically targeting NHE3, tenapanor reduces luminal sodium uptake in the gut, with greater than 100-fold selectivity over other isoforms such as NHE1 and NHE2 (IC₅₀ for human NHE3: 13 nM; IC₅₀ for human NHE1 and NHE2: >10 μM). This inhibition results in proton retention within epithelial cells, causing intracellular acidification that modulates proteins, thereby decreasing paracellular permeability. The localized action in the minimizes effects on renal NHE3 due to tenapanor's poor oral and negligible systemic , with concentrations typically below the limit of quantification (<0.5 ng/mL) after oral dosing. In the context of irritable bowel syndrome with constipation (IBS-C), NHE3 inhibition by tenapanor decreases sodium-dependent fluid absorption, leading to retention of water and sodium in the intestinal lumen, which increases stool frequency and consistency through enhanced luminal fluid content. For hyperphosphatemia in chronic kidney disease (CKD), the same mechanism reduces paracellular phosphate flux across the intestinal epithelium by tightening tight junctions, thereby limiting dietary phosphate absorption without relying on systemic exposure.

Pharmacokinetics

Tenapanor exhibits minimal systemic absorption following oral administration, with plasma concentrations of the parent drug typically below the limit of quantitation (less than 0.5 ng/mL) in the majority of samples from healthy subjects after single or repeated doses of 50 mg twice daily. This low absorption is attributed to its mechanism of , which confines its action primarily to the gastrointestinal tract. Peak plasma levels of the major metabolite M1 occur approximately 1 hour post-dose, reaching about 13 ng/mL after a single 50 mg dose and 15 ng/mL at steady state. Distribution of tenapanor is limited due to its poor absorption, with the drug primarily acting locally in the gut and minimal penetration into systemic circulation. In vitro, tenapanor and its major metabolite M1 are highly bound to plasma proteins, approximately 99% and 97%, respectively. The volume of distribution has not been precisely determined owing to the drug's low systemic exposure. Tenapanor undergoes metabolism primarily by CYP3A4 and CYP3A5 enzymes in the liver and gut, producing low levels of the major metabolite M1, which is detected in plasma but does not contribute significantly to pharmacological activity. No active metabolites have been identified. Elimination of tenapanor occurs predominantly via fecal excretion, with approximately 70% of a radiolabeled dose recovered in feces within 120 hours post-dose (and 79% within 240 hours), mostly as unchanged parent drug (65% of the dose within 144 hours). Urinary excretion accounts for about 9% of the dose, primarily as metabolites, with less than 5% as unchanged drug. The half-life of the parent drug cannot be reliably determined due to its minimal systemic absorption, while the metabolite M1 has a half-life of approximately 19 hours in humans. In special populations, no dose adjustment is required for patients with mild to moderate hepatic impairment, as plasma concentrations of tenapanor remain below the limit of quantitation, and exposure to M1 is reduced by about 27-33% compared to healthy subjects. Similarly, no adjustment is needed for renal impairment, including end-stage renal disease, where M1 concentrations are comparable or slightly lower than in healthy individuals. The effects of dialysis on tenapanor pharmacokinetics have not been specifically studied.

Chemistry

Structure and Properties

Tenapanor is a synthetic small molecule characterized by the molecular formula C<sub>50</sub>H<sub>66</sub>Cl<sub>4</sub>N<sub>8</sub>O<sub>10</sub>S<sub>2</sub> for its form. Its molecular weight is 1145.04 g/mol. The compound features a symmetric consisting of two (4S)-6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide units linked via a central moiety and poly()-like chains, conferring at two stereocenters. The IUPAC name for tenapanor is 1-[2-[2-[2-[[3-[(4S)-6,8-dichloro-2-methyl-3,4-dihydro-1H-isoquinolin-4-yl]phenyl]sulfonylamino]ethoxy]ethoxy]ethyl]-3-[4-[2-[2-[2-[[3-[(4S)-6,8-dichloro-2-methyl-3,4-dihydro-1H-isoquinolin-4-yl]phenyl]sulfonylamino]ethoxy]ethoxy]ethylcarbamoyl]phenyl]. In its salt form, used in pharmaceutical formulations, the is C<sub>50</sub>H<sub>68</sub>Cl<sub>6</sub>N<sub>8</sub>O<sub>10</sub>S<sub>2</sub> with a molecular weight of 1218 . Tenapanor appears as a white to off-white to light brown hygroscopic . It exhibits poor , with values as low as 0.0000595 mg/mL at 6.8, classifying it as a (BCS) Class IV compound. The computed value of approximately 5.1 indicates significant . This low aqueous supports its as oral tablets, where it is minimally absorbed systemically. Regarding stability, tenapanor maintains integrity for a 24-month expiration period when stored at in (HDPE) bottles with , ensuring suitability for solid oral . Its hygroscopic nature necessitates controlled humidity during handling and storage to prevent degradation.

Synthesis

The synthesis of tenapanor, a dimeric , involves a multi-step to construct the core, followed by formation with a poly(ethylene glycol) side chain, and final dimerization via a butane-1,4-diyl linker. The initial discovery route, as described in U.S. No. 8,969,377, begins with the preparation of the core scaffold from 1-(3-bromophenyl)ethanone. Bromination yields 2-bromo-1-(3-bromophenyl)ethanone, which reacts with (2,4-dichlorophenyl)-N-methylmethanamine in and triethylamine to form 1-(3-bromophenyl)-2-((2,4-dichlorobenzyl)(methyl)amino)ethanone. Reduction with in produces the corresponding alcohol, which undergoes acid-catalyzed cyclization with to afford 4-(3-bromophenyl)-6,8-dichloro-2-methyl-1,2,3,4-. Substitution with phenylmethanethiol, followed by oxidation, generates the sulfonyl chloride intermediate, 3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzene-1-. This is coupled with 2-(2-(2-aminoethoxy)ethoxy)ethanamine to form the monomer, (S)-N-(2-(2-(2-aminoethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-methyl-1,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide (referred to as (S)-Compound A), with achieved during the cyclization step to favor the (S) configuration at the 4-position. The dimerization couples two equivalents of (S)-Compound A with 1,4-diisocyanatobutane to form the urea-linked tenapanor , yielding approximately 36.7% overall with impurities necessitating () purification. Optimization for scale-up addressed the low yield and scalability issues of the discovery route by replacing the toxic and non-selective 1,4-diisocyanatobutane with safer alternatives, as detailed in Ardelyx's WO2020051014A1 patent. One improved route reacts (S)-Compound A with N,N'-(butane-1,4-diyl)bis(1H-imidazole-1-carboxamide), prepared from and 1,4-diaminobutane, in (15-20 volumes) at 50-55°C, achieving 85-90% and 96-98% purity for the tenapanor . An alternative route involves sequential reaction of (S)-Compound A with to form the imidazolyl carboxamide intermediate (not isolated), followed by addition of 1,4-diaminobutane in (15-20 volumes) at 40-50°C, yielding 85-90% with 95-96% purity while maintaining through the chiral (S)-Compound A. Key intermediates, such as the NHE3-binding motifs in (S)-Compound A, are purified via from //water (15:5:2 volumes) at 50-55°C, resulting in 99.5% purity suitable for kilogram-scale production. These routes avoid hazardous reagents, enhance , and enable industrial scalability without extensive . For , the tenapanor is converted to the dihydrochloride salt by treatment with (2-2.1 molar equivalents) in at 0-5°C, yielding a crystalline form with powder diffraction peaks at 11.1°, 21.1°, 21.6°, 22.7°, and 24.0° 2θ, and low levels of (5.9-6.1% w/w) and hydroxy impurities (<0.02% w/w). This salt form facilitates tablet production due to its stability and handling properties.

Development and Clinical Trials

Discovery and Preclinical Studies

Ardelyx, Inc., founded in 2007, identified the sodium/hydrogen exchanger isoform 3 (NHE3) as a key target for addressing gastrointestinal disorders due to its central role in intestinal sodium absorption and fluid homeostasis. The company's early research, spanning 2007 to 2012, emphasized developing gut-restricted to modulate sodium transport locally without systemic exposure, aiming to treat conditions like constipation and phosphate dysregulation in . Lead optimization efforts began with high-throughput screening of tetrahydroisoquinoline (THIQ) compounds using a cell-based deacidification assay in NHE3-transfected opossum kidney cells, identifying initial hits with moderate potency. Subsequent medicinal chemistry focused on dimeric macrocyclic structures incorporating polyethylene glycol linkers to enhance selectivity and minimize absorption, culminating in tenapanor (also known as RDX5791 or AZD1722), a potent NHE3 inhibitor with IC<sub>50</sub> values of 5 nM () and 10 nM (). This optimization prioritized the (S,S)-stereoisomer configuration for optimal binding and pharmacokinetic properties, enabling localized action in the gut. Preclinical pharmacodynamic studies in rats demonstrated tenapanor's , with oral dosing at 10 mg/kg increasing stool water content by 56% and elevating fecal sodium and levels while reducing urinary of these electrolytes, confirming reduced intestinal absorption without altering systemic sodium balance. In dogs and polarized Madin-Darby canine cell models, tenapanor exhibited low permeability (P<sub>app</sub> < 0.2 × 10<sup>−6</sup> cm/s), supporting its minimal systemic and gut-specific effects. These models highlighted tenapanor's ability to promote fecal water and , establishing proof-of-concept for its therapeutic potential in sodium- and phosphate-related disorders. Toxicology assessments, including a seven-day repeat-dose study in rats, revealed no significant organ or , as evidenced by negative results in the and minimal inhibition (<50% at 10 μM). Gastrointestinal-specific adverse effects, such as diarrhea and dehydration, were observed at high doses (≥30 mg/kg), consistent with exaggerated pharmacology but resolving without long-term sequelae. Initial patents covering tenapanor's composition were filed by in 2010, providing intellectual property protection extending to 2029. Preceding clinical advancement, explored partnerships, culminating in a 2012 worldwide licensing agreement with for tenapanor and related NHE3 inhibitors, which included upfront payments and milestones to support further development.

Phase 3 Trials for IBS-C

The pivotal Phase 3 clinical trials for tenapanor in irritable bowel syndrome with constipation (IBS-C), T3MPO-1 and T3MPO-2, were randomized, double-blind, placebo-controlled studies evaluating the drug's efficacy and safety in adults meeting Rome III criteria for IBS-C. T3MPO-1 enrolled 606 patients across 92 U.S. sites for a 12-week treatment period followed by a 4-week randomized withdrawal phase, with participants randomized 1:1 to tenapanor 50 mg twice daily (n=307) or placebo (n=299) in the intention-to-treat population. The primary endpoint was the proportion of overall responders achieving both a ≥30% reduction from baseline in average weekly worst abdominal pain and an increase of ≥1 complete spontaneous bowel movement (CSBM) per week in the same week, for at least 6 of the first 12 treatment weeks. Tenapanor met this endpoint, with 27.0% of patients responding compared to 18.7% on placebo (P=0.020). Key secondary endpoints included CSBM responders (≥1 CSBM increase for ≥6/12 weeks) and abdominal pain responders (≥30% pain reduction for ≥6/12 weeks), showing 33.9% vs. 29.4% (P=0.27) for CSBM and 44.0% vs. 33.1% (P=0.008) for pain relief. A stricter analysis for durable response (≥9/12 weeks on the composite endpoint) further supported efficacy, with 13.7% vs. 3.3% (P<0.001). T3MPO-2 involved 620 patients in a 26-week treatment period to assess longer-term efficacy, with randomization to tenapanor 50 mg twice daily (n=293) or placebo (n=300) in the intention-to-treat population across multiple U.S. sites. The primary endpoint mirrored T3MPO-1, focusing on the first 12 weeks, and was achieved by 36.5% of tenapanor patients versus 23.7% on placebo (P<0.001). Efficacy persisted through week 26, with significant improvements in secondary endpoints such as CSBM frequency (mean increase of 1.7 vs. 0.9 weekly CSBMs, P<0.001) and abdominal pain reduction (P<0.05). The trial included a 4-week withdrawal period to evaluate symptom rebound, confirming sustained benefits without notable worsening upon discontinuation.
TrialPrimary Endpoint Responder Rate (Tenapanor vs. Placebo)P-valueDuration
T3MPO-127.0% vs. 18.7%0.02012 weeks
T3MPO-236.5% vs. 23.7%<0.00126 weeks
Subgroup analyses from both trials demonstrated consistent efficacy across demographics, including age (≤65 vs. >65 years), sex (male vs. female), and categories, though smaller subgroup sizes limited statistical power for some comparisons. For instance, the composite endpoint showed favorable trends in women (predominant in the cohorts at ~81%) and across levels, with no significant interactions identified. The T3MPO-3 open-label extension study evaluated long-term safety in 312 patients rolling over from T3MPO-1 or T3MPO-2, administering tenapanor 50 mg twice daily for up to 52 weeks (additional 39 weeks post-T3MPO-1 or 26 weeks post-T3MPO-2). Of these, 84% (262 patients) completed the study, with 90 receiving ≥52 weeks of treatment. Safety was consistent with shorter trials, focusing on tolerability rather than efficacy endpoints. Across the trials, adverse events were primarily gastrointestinal, with diarrhea occurring in 14.8% of tenapanor patients versus 2.3% on ; this led to discontinuation in 6.8% versus 0.8%. No non-inferiority assessments were performed, and statistical analyses used Cochran-Mantel-Haenszel tests adjusted for baseline factors, confirming the trials' rigor without evidence of imbalance. These results established tenapanor's role in improving CSBM frequency and relief in IBS-C, leveraging its mechanism as a sodium/ exchanger 3 to enhance intestinal fluid secretion.

Phase 3 Trials for

The pivotal Phase 3 trials evaluating tenapanor for the of in patients with on demonstrated its efficacy in reducing serum levels through inhibition of intestinal sodium/ exchanger 3 (NHE3), thereby limiting paracellular in the gut. The ESRD trial (TEN-02-201) was a randomized, double-blind, -controlled study involving 219 patients on maintenance with (serum ≥5.5 mg/dL). It featured an 8-week period followed by a 4-week randomized period, assessing tenapanor at doses of 3 mg, 10 mg, or 30 mg twice daily (titrated as needed) versus . The primary endpoint was the mean change in serum during the period, showing -0.04 mg/dL for tenapanor versus +0.65 mg/dL for (p<0.001). A secondary responder showed approximately 49% of tenapanor-treated patients achieved a ≥1.2 mg/dL reduction from baseline during , compared to 24% on . was performed on the intent-to-treat population, with no cardiovascular safety signals observed, as serious adverse events were comparable between groups and none were attributed to cardiovascular causes. The confirmatory PHREEDOM trial (TEN-02-301) was a 52-week, phase 3 study in 564 patients on maintenance with , consisting of a 26-week open-label tenapanor (doses titrated from 10 mg to 30 mg twice daily) followed by a 12-week double-blind, placebo-controlled randomized . The primary was the change in during the , met with an increase of 0.7 mg/dL (95% CI: 0.2, 1.1; p=0.002) in the group versus continued tenapanor. Approximately 68% of participants maintained reductions below baseline throughout the open-label phase, confirming long-term efficacy and safety without new concerns. No cardiovascular safety signals were observed. The AMPLIFY trial (TEN-02-202) was a 4-week, randomized, double-blind, placebo-controlled study of tenapanor (30 mg twice daily) as add-on therapy to existing phosphate binders in 236 patients on maintenance dialysis with inadequately controlled hyperphosphatemia (serum phosphorus ≥5.5 mg/dL and ≤10.0 mg/dL). The primary endpoint was the change in serum phosphorus from baseline to week 4, showing a greater reduction with tenapanor plus binders (-0.84 mg/dL) versus placebo plus binders (-0.19 mg/dL; p<0.001). A key secondary endpoint, the proportion of responders achieving serum phosphorus <5.5 mg/dL, was met by 49.2% of tenapanor-treated patients versus 23.4% on placebo (p<0.001), analyzed via ITT with no cardiovascular safety signals reported. Subgroup analyses revealed a greater treatment effect in patients intolerant to phosphate binders, where tenapanor enabled improved phosphorus control; additionally, binder dose reductions were observed in 27% of tenapanor recipients, supporting its role in reducing pill burden.

Regulatory Approvals

United States

Tenapanor, marketed under the brand name IBSRELA by Ardelyx, Inc., received U.S. (FDA) approval on September 12, 2019, for the treatment of with (IBS-C) in adults 18 years and older. The (NDA 211801) was submitted on September 12, 2018, and the approval was based on data from the phase 3 T3MPO-1 and T3MPO-2 trials, which showed statistically significant improvements in 12-week abdominal and bowel symptoms compared to . Ardelyx holds marketing exclusivity for this indication until 2026. For the treatment of hyperphosphatemia in (CKD), tenapanor, branded as XPHOZAH, underwent a more protracted approval process. Ardelyx submitted the initial (213931) on June 29, 2020, but received a complete response letter on July 28, 2021, due to FDA concerns regarding the small magnitude of the treatment effect on phosphorus levels. After appealing the decision, providing additional analyses, and receiving favorable input from an FDA advisory committee in 2022, Ardelyx resubmitted the on April 17, 2023. The FDA approved XPHOZAH on October 17, 2023, as an add-on therapy to phosphate binders for reducing phosphorus in adults with CKD on , supported by phase 3 trial results demonstrating a mean reduction of 0.72 mg/dL in phosphorus. Post-approval label updates have addressed considerations for both indications. In , the prescribing information was revised to include warnings about potential drug interactions, noting that tenapanor inhibits the intestinal OATP2B1 transporter, which may reduce absorption and efficacy of certain medications such as statins (e.g., ) and enalapril. In 2025, the labels for IBSRELA and XPHOZAH were updated to reinforce the in children under 6 years of age, citing preclinical studies in juvenile rats that showed mortality due to severe and . Ardelyx is fulfilling FDA post-marketing commitments, including ongoing long-term studies to assess risks such as and in CKD patients on .

Canada and Other Regions

In Canada, Health Canada approved tenapanor, marketed as Ibsrela, for the treatment of irritable bowel syndrome with constipation (IBS-C) in adults on April 17, 2020. As of November 2025, tenapanor remains under review for the indication by Health Canada through Knight Therapeutics, with no approval granted to date. In the , tenapanor has not received centralized marketing authorization from the as of 2025, though it was under review for in prior years; national-level applications remain pending in select member states. Availability in the EU is thus limited, with access primarily through individual country compassionate use programs or named-patient imports. Outside and , tenapanor received approval in for the treatment of in patients with on on September 25, 2023, following positive Phase 3 results; Kyowa Kirin, Ardelyx's partner, launched the product as PHOZEVEL in February 2024. In , tenapanor (IBSRELA) was approved for IBS-C on September 29, 2023, by the Department of Health through . In , tenapanor was approved on February 26, 2025, for in CKD patients on under the brand name Wan Ti Le by . In and , tenapanor is not fully approved by the or Medsafe, respectively, and access is restricted to compassionate use or special access schemes for eligible patients. Pricing for Ibsrela in Canada is approximately CAD 350 per month for a standard 50 mg twice-daily regimen (60 tablets), which is lower than comparable U.S. costs but presents access barriers due to limited public reimbursement, particularly for off-label or unapproved uses like chronic kidney disease-related hyperphosphatemia. Reimbursement challenges persist for the CKD indication across regions, stemming from regulatory delays and the need for demonstrated cost-effectiveness in national health systems.

Adverse Effects

Common Side Effects

The most common adverse reaction associated with tenapanor across clinical trials is diarrhea, resulting from its inhibition of the sodium-hydrogen exchanger 3 (NHE3) in the gastrointestinal tract, which increases fluid secretion into the intestinal lumen. In two phase 3 trials for irritable bowel syndrome with constipation (IBS-C), diarrhea occurred in 15-16% of patients receiving tenapanor 50 mg twice daily compared to 2-4% on placebo, with severe cases in 2.5% versus 0.2%; symptoms were typically mild to moderate and onset within 1-2 weeks. In phase 3 trials for hyperphosphatemia in chronic kidney disease patients on dialysis, the incidence was higher at 43-53% for the same dose, with severe diarrhea in approximately 5%, often dose-dependent in earlier studies but consistent at the approved regimen. Other gastrointestinal effects include and , reported in 2-3% and 3% of IBS-C trial patients, respectively, versus less than 1% on ; these were generally transient and mild. occurred in about 2% of IBS-C patients compared to less than 1% on . Discontinuation rates due to gastrointestinal effects ranged from 7-9% in IBS-C trials and up to 16% in trials, primarily attributed to . In cases of overdose, symptoms are expected to mirror exaggerated therapeutic effects, with severe potentially leading to and imbalances such as , though no specific human overdose data exist; management involves supportive care including fluid and replacement.

Contraindications and Precautions

Tenapanor is contraindicated in pediatric patients younger than of age, as administration to young juvenile rats resulted in deaths presumed to be due to following doses of 10 mg/kg/day or greater. It is also contraindicated in patients with known or suspected mechanical gastrointestinal obstruction, due to the potential for exacerbation of the condition. No dosage adjustment of tenapanor is recommended for patients with mild or moderate hepatic impairment; the of tenapanor in patients with severe hepatic impairment have not been studied. Use during lacks established data in humans, with no assigned; animal reproduction studies revealed no direct or indirect harmful effects on embryofetal development at exposures up to 15 times the recommended human dose. No dosage adjustment is required for patients with (CKD), including those on ; however, volume status should be monitored in patients due to the risk of from . Drug interactions include reduced absorption of organic anion transporting polypeptide 2B1 (OATP2B1) substrates (e.g., enalapril), with decreases in Cmax of approximately 70% and of 50-65%, warranting monitoring and potential dose increases of the substrate. A warning exists for the risk of severe occurring in less than 6% of adults, which may lead to ; patients should maintain adequate , and dosing should be suspended with rehydration if severe, potentially resuming at a reduced dose (e.g., 25 mg twice daily for IBS-C or 15 mg twice daily for ) upon resolution.