1260643-52-4
Hyperkalemia
Patiromer is a non-absorbed, cation exchange polymer developed for the oral treatment of hyperkalemia, a condition characterized by elevated potassium levels in the blood.[1] Marketed under the brand name Veltassa, it represents a significant therapeutic advancement, being one of the first new medications approved for this indication in over five decades.[1] This development addresses a long-standing clinical need for effective and well-tolerated agents to manage chronic hyperkalemia, particularly as an alternative to older resins like sodium polystyrene sulfonate, which are often associated with poor tolerability and safety concerns.[4]
Patiromer is classified as a small molecule potassium binder and is specifically indicated for the management of hyperkalemia in adults and pediatric patients aged 12 years and older.[2] A critical limitation of its use, however, is that it is not intended for the emergency treatment of life-threatening hyperkalemia due to its delayed onset of action, which is approximately seven hours.[1] Acute, severe hyperkalemia requires immediate interventions such as intravenous calcium, insulin and glucose infusions, or hemodialysis to rapidly stabilize cardiac cell membranes and shift potassium intracellularly.[1] Patiromer's role is therefore firmly established in the chronic or sub-acute management of elevated potassium levels, where sustained control is the primary therapeutic goal.[4]
The primary clinical value of patiromer extends beyond simply lowering serum potassium; it functions as a crucial "enabling therapy." This is particularly relevant for patients with chronic kidney disease (CKD) and heart failure (HF), for whom treatment with renin-angiotensin-aldosterone system inhibitors (RAASi)—such as angiotensin-converting enzyme (ACE) inhibitors, angiotensin II receptor blockers (ARBs), and mineralocorticoid receptor antagonists (MRAs)—is a cornerstone of guideline-directed medical therapy.[10] While RAASi therapies are proven to reduce morbidity and mortality in these populations, their use is frequently limited or necessitated for discontinuation by the development of hyperkalemia, a common and predictable side effect.[10]
By effectively binding and removing potassium from the gastrointestinal tract, patiromer allows clinicians to initiate or maintain patients on optimal, life-saving doses of RAASi, thereby addressing the underlying cardiorenal disease more aggressively.[10] This RAASi-enabling capacity is a central component of its therapeutic profile. Furthermore, patiromer is a sodium-free formulation that exchanges calcium for potassium, a key differentiator from sodium-based binders that can contribute to fluid retention and exacerbate hypertension or heart failure.[13] Its mechanism of action is localized to the gut, leading to increased fecal potassium excretion without systemic absorption, which contributes to a predictable safety profile.[2]
For clarity and precision in clinical and research settings, a comprehensive list of identifiers for patiromer is essential. The drug is available in a formulated complex, and distinguishing between the active moiety and its clinical form is critical for accurate documentation and communication.
Identifier | Value | Source(s) |
---|---|---|
Generic Name (INN) | Patiromer | 2 |
Brand/Trade Names | Veltassa, RLY5016 | 1 |
Type/Modality | Small Molecule, Macromolecular Substance | 2 |
Drug Class | Potassium Binder, Cation Exchange Resin, Potassium-Removing Agent | 2 |
Active Moiety | Patiromer | 15 |
Formulated Complex | Patiromer Sorbitex Calcium | 1 |
CAS Number (Free Form) | 1260643-52-4 | 1 |
CAS Number (Calcium Salt) | 1208912-84-8 | 1 |
CAS Number (Sorbitex Calcium) | 1415477-49-4 | 18 |
DrugBank ID | DB09263 | 1 |
UNII | 1FQ2RY5YHH | 1 |
ATC Code | V03AE09 | 1 |
KEGG ID | D10148 (Patiromer), D11037 (Calcium Salt) | 1 |
PubChem SID | 135626866 | 1 |
RXCUI | 1716203 | 15 |
Patiromer is a synthetic, high-molecular-weight polymer that is not defined by a simple chemical formula but rather by its constituent monomers and cross-linking agents.[1] The active polymer is synthesized from three primary components: 2-fluoroacrylic acid, which provides the carboxylate groups for cation exchange; divinylbenzenes (both ortho- and para-isomers), which create a cross-linked network structure; and 1,7-octadiene, which acts as an additional cross-linker to ensure the polymer's integrity and insolubility.[1]
The substance used in clinical practice is not the free polymer but a specifically formulated complex known as patiromer sorbitex calcium.[1] This complex consists of the active patiromer polymer, where the acidic protons of the fluoroacrylic acid units are replaced with calcium ions (
Ca2+), and D-sorbitol, which serves as a counterion complex.[2] The ratio of calcium to fluoroacrylic acid units is 1:2, and there is one molecule of sorbitol per two calcium ions.[1] Consequently, the formal chemical name for the drug substance is "cross-linked polymer of calcium 2-fluoroprop-2-enoate with diethenylbenzene and octa-1,7-diene, combination with D-glucitol".[2] Each gram of the active moiety, patiromer, is equivalent to a nominal amount of 2 grams of the patiromer sorbitex calcium complex.[2]
Patiromer sorbitex calcium is an off-white to light brown, amorphous, free-flowing powder.[1] A key physical characteristic derived from its manufacturing process is its composition of individual, insoluble spherical beads of a uniform and controlled size.[17] This morphology is a deliberate design feature. The polymer is fundamentally insoluble in common solvents, including water, 0.1 M hydrochloric acid, n-heptane, and methanol.[1] This insolubility is critical to its mechanism of action, ensuring that it remains within the gastrointestinal lumen and is not systemically absorbed.[17]
For clinical use, patiromer is formulated as a powder for oral suspension, packaged in single-use sachets containing various doses of the active moiety, typically 8.4 g, 16.8 g, or 25.2 g.[22] The formulation also contains the inactive ingredient xanthan gum, which likely serves as a suspending agent to improve the consistency and palatability of the mixture when reconstituted.[25]
The specific chemical and physical design of patiromer is directly linked to its function and improved tolerability profile compared to older potassium binders. The manufacturing process, which utilizes suspension polymerization, results in uniform, spherical beads with a median particle size of approximately 118 micrometers, with 90% of particles falling within the 74 to 179 micrometer range.[21] This contrasts sharply with the physical form of first-generation resins like sodium polystyrene sulfonate (SPS), which consist of irregular, jagged-shaped crystal fragments with a broad and uncontrolled particle size distribution.[21]
This structural difference is not merely aesthetic but has significant clinical implications. The older resins, particularly SPS, are associated with poor gastrointestinal (GI) tolerability and, in rare but serious cases, complications such as colonic necrosis.[5] While the etiology of such complications is likely multifactorial, the physical irritation of the GI mucosa by sharp, irregular particles may be a contributing factor. The development of patiromer with smooth, spherical beads can be understood as a deliberate pharmacotechnical advancement aimed at minimizing direct physical abrasion of the intestinal lining. This refined morphology is thought to contribute to its more favorable GI safety profile, which is a prerequisite for a medication intended for chronic, long-term administration. This structural superiority allows patients to remain on therapy for extended periods, which is essential for the continuous management of hyperkalemia and the enabling of RAASi therapy.
Patiromer exerts its therapeutic effect through a straightforward physicochemical mechanism of action within the gastrointestinal (GI) tract.[2] It is a non-absorbed, high-capacity, synthetic cation exchange polymer that operates by binding potassium ions (
K+) present in the intestinal lumen.[2] The polymer is formulated with calcium (
Ca2+) as its counterion.[1] When administered orally, patiromer travels through the GI tract, where it exchanges its calcium ions for free potassium ions from the intestinal contents.[1]
This exchange process occurs throughout the GI tract but is optimized to be most efficient in the distal colon.[5] The colon is the primary site of action for two key reasons: it is where the concentration of free potassium is highest, and the transit time is longest, allowing for maximal binding to occur.[5] The polymer was specifically designed to be fully ionized at the physiological pH of the colon, which further enhances the efficiency of the ion exchange.[5] The bound potassium remains trapped within the polymer matrix, which is then excreted from the body via the feces.[2] This process effectively intercepts potassium that would otherwise be absorbed into the bloodstream, leading to a net increase in fecal potassium excretion and a corresponding reduction in serum potassium levels.[1]
The pharmacodynamic effects of patiromer are characterized by a relatively slow but sustained reduction in serum potassium. The onset of action, defined as a detectable decrease in serum potassium levels, occurs approximately seven hours after the first dose is administered.[1] This delayed onset underscores why patiromer is not suitable for emergency situations requiring rapid potassium correction.[1]
Following the initial dose, if treatment is continued, serum potassium levels continue to decline for at least 48 hours, eventually reaching a new, lower steady state.[1] After the administration of the final dose, the potassium-lowering effect is maintained for approximately 24 hours, which supports the clinical practice of once-daily dosing.[1] This pharmacodynamic profile is well-suited for the chronic management of hyperkalemia, providing consistent, around-the-clock control of potassium levels with a convenient dosing schedule.
The pharmacokinetic profile of patiromer is exceptionally simple and predictable due to its fundamental nature as a non-absorbed polymer.
This lack of systemic absorption is the central pillar of patiromer's safety and predictability. By confining its pharmacological activity and primary adverse effects to the GI tract, it eliminates the risks of systemic off-target effects, hepatic metabolism, or renal clearance that complicate the profiles of many systemically absorbed drugs. This is a particularly advantageous feature for its target patient population, which often includes individuals with severe CKD and compromised drug clearance capabilities. The drug's behavior is therefore highly predictable, regardless of a patient's renal or hepatic function, simplifying its clinical application in complex, polymedicated individuals. This "pharmacokinetic simplicity" represents a major safety advantage over systemically active medications.
The clinical development program for patiromer was strategically designed to establish its efficacy for both acute reduction and long-term maintenance of normokalemia, and to prove its value as an enabling therapy for RAAS inhibitors. The two pivotal studies, AMETHYST-DN (Phase 2) and OPAL-HK (Phase 3), provide a complementary and robust evidence base for its clinical use. The sequential nature of these trials reflects a deliberate pathway: AMETHYST-DN first established long-term safety and identified effective dosing ranges, which was essential for a chronic-use medication. Following this, the more methodologically rigorous OPAL-HK trial was designed to provide unequivocal, placebo-controlled evidence of patiromer's ability to maintain normokalemia and, critically, to prove the RAASi-enabling hypothesis required for regulatory approval and broad clinical acceptance. This logical progression from long-term safety to robust, placebo-controlled efficacy demonstrates a well-conceived clinical development strategy.
The OPAL-HK trial was a landmark Phase 3 study that definitively established the efficacy of patiromer.[29] It enrolled patients with CKD who were on stable RAASi therapy and presented with serum potassium levels between 5.1 and <6.5 mmol/L.[10] The trial featured an innovative two-part design. Part A was a 4-week, single-arm, open-label initial treatment phase where all participants received patiromer to lower their serum potassium. Part B was an 8-week, randomized, double-blind, placebo-controlled withdrawal phase. Only patients who had moderate to severe hyperkalemia at baseline and achieved normokalemia (serum K+ 3.8 to <5.1 mmol/L) at the end of Part A were eligible to be randomized in Part B to either continue patiromer or switch to a placebo.[10] This withdrawal design is a powerful method to demonstrate that a drug is actively maintaining a therapeutic effect; if the condition returns upon withdrawal, it confirms the drug's efficacy.
The results from OPAL-HK were compelling. In Part A, treatment with patiromer resulted in a statistically significant mean reduction in serum potassium of -1.01 mmol/L from baseline (p<0.001).[29] By the end of the 4-week treatment phase, 76% of patients had successfully reached the target normokalemic range.[29] Subgroup analyses confirmed that this efficacy was consistent in older adults (≥65 years), a key demographic for this condition.[32]
The results of Part B were equally definitive. Patients who continued on patiromer maintained stable, normal potassium levels. In stark contrast, those who were switched to placebo experienced a rapid and significant increase in their serum potassium.[29] The recurrence of hyperkalemia, defined as a potassium level of ≥5.5 mmol/L, occurred in only 15% of patients in the patiromer group compared to 60% of patients in the placebo group (p<0.001).[29] In the older adult subgroup, recurrence of hyperkalemia (defined as K+ ≥5.1 mEq/L) was observed in 30% of patiromer-treated patients versus 92% of those on placebo, demonstrating a profound preventive effect.[32]
A critical secondary outcome of the OPAL-HK trial was its demonstration of patiromer's role in facilitating guideline-directed medical therapy. An exploratory analysis revealed that at the end of the 12-week study period, 94% of patients who received patiromer throughout both phases were able to remain on their RAASi therapy.[30] This was in sharp contrast to the placebo group, where only 44% of patients could continue their RAASi therapy due to recurrent hyperkalemia.[30] This finding provides strong evidence for patiromer's value as an enabling agent, allowing high-risk cardiorenal patients to receive the full, life-prolonging benefits of their RAASi medications.
The AMETHYST-DN trial was a 52-week, multicenter, open-label, Phase 2 study designed to evaluate the long-term safety and efficacy of various starting doses of patiromer.[10] The trial enrolled 306 patients with type 2 diabetes, CKD (eGFR 15 to <60 mL/min/1.73 m2), and hyperkalemia (serum K+ >5.0 mEq/L), all of whom were receiving RAASi therapy.[34] Participants were stratified based on the severity of their baseline hyperkalemia—mild (K+ >5.0 to 5.5 mEq/L) or moderate (K+ >5.5 to <6.0 mEq/L)—and then randomized to one of several starting dose groups, with doses ranging from 8.4 g/day to 33.6 g/day.[33] Doses were then titrated as needed to maintain serum potassium within the target range.
AMETHYST-DN successfully demonstrated that patiromer provides durable control of serum potassium over an extended period. All starting doses across both strata produced statistically significant reductions in serum potassium by week 4, and these reductions were effectively maintained for the entire 52-week duration of the study.[33] The potassium-lowering effect was evident early, with mean potassium levels showing a significant reduction by day 3 of treatment.[36] Importantly, the efficacy was consistent across different age cohorts, including patients aged 75 years and older, confirming its utility in the elderly population.[36] The study provided crucial data on the long-term safety and tolerability of patiromer, paving the way for the pivotal Phase 3 trial.
Parameter | OPAL-HK | AMETHYST-DN |
---|---|---|
Study Name | OPAteL in HyperKalemia | A Multicenter, Randomized, Open-Label, Dose Ranging Study to Evaluate the Efficacy and Safety of Patiromer in the Treatment of Hyperkalemia in Patients With Hypertension and Diabetic Nephropathy |
Trial Phase | Phase 3 | Phase 2 |
Study Design | Two-part: 4-week open-label treatment phase, followed by an 8-week randomized, placebo-controlled withdrawal phase. | 52-week, open-label, randomized, dose-ranging. |
Patient Population | Patients with CKD on RAASi therapy with serum K+ 5.1 to <6.5 mmol/L. | Patients with Type 2 Diabetes, CKD (eGFR 15-<60), and hyperkalemia (K+ >5.0 mEq/L) on RAASi therapy. |
Sample Size (n) | 243 (Part A), 107 (Part B) | 306 |
Duration | 12 weeks | 52 weeks |
Primary Efficacy Endpoint | Part A: Mean change in serum K+ at Week 4. Part B: Difference in serum K+ change between patiromer and placebo. | Mean change in serum K+ from baseline to Week 4. |
Key Efficacy Results | Part A: Mean K+ reduction of -1.01 mmol/L (p<0.001). Part B: Recurrence of hyperkalemia (K+ ≥5.5) in 15% (patiromer) vs. 60% (placebo) (p<0.001). | Statistically significant dose-dependent reductions in serum K+ at Week 4, which were sustained for 52 weeks. |
Key Safety Findings | Most common adverse event was mild-to-moderate constipation (11%). Hypokalemia occurred in 3%. | Most common treatment-related adverse event was hypomagnesemia (7.2%). Constipation occurred in 6.3%. |
Key Conclusion/Implication | Patiromer effectively lowers serum K+ and significantly reduces the recurrence of hyperkalemia, enabling continued RAASi therapy. | Patiromer provides safe and durable long-term control of serum K+ in patients with diabetic kidney disease. |
The safety profile of patiromer has been well-characterized through its clinical trial program, with adverse events being predominantly gastrointestinal and generally mild to moderate in severity.[13] As a non-absorbed polymer, its effects are localized to the gut, which is reflected in its adverse event profile.
Adverse Reaction | Incidence (%) in Clinical Trials | Clinical Comment/Monitoring Recommendation |
---|---|---|
Constipation | 7.2% | The most common adverse event. Patients should be counseled on maintaining adequate hydration. Use with caution in patients with a history of severe constipation. |
Hypomagnesemia | 5.3% | Due to binding of magnesium in the colon. Serum magnesium should be monitored, particularly during the first month of therapy, and supplemented if necessary. |
Diarrhea | 4.8% | Generally mild to moderate. |
Nausea | 2.3% | Can be managed symptomatically. |
Abdominal Discomfort | 2.0% | Generally mild and transient. |
Flatulence | 2.0% | Common GI side effect. |
Hypokalemia | 3.0% - 5.6% | Risk of overcorrection. Regular monitoring of serum potassium is essential to guide dose titration and prevent levels from falling below the target range. |
Hypersensitivity Reactions | 0.3% | Rare, but patients should be aware of signs such as swelling of the lips, face, or throat. Discontinue if a reaction occurs. |
The prescribing information for patiromer includes several important warnings and precautions to ensure its safe use:
Data gathered from global pharmacovigilance databases since patiromer's approval have provided valuable real-world insights into its safety profile. Analysis of several years of post-marketing data has confirmed that the tolerability and safety of patiromer in routine clinical practice are predictable and consistent with the findings from the pivotal clinical trials.[12] Importantly, this surveillance has not identified any new or unexpected safety signals, reinforcing the well-characterized risk-benefit profile of the medication.[12]
The potential for drug-drug interactions with patiromer stems directly from its mechanism of action. As a non-absorbed, non-specific cation exchange polymer, it has the capacity to bind to other orally administered medications within the GI tract, in addition to its target, potassium.[2] This binding can sequester the co-administered drug, preventing its absorption from the gut into the bloodstream. The clinical consequence of this interaction is a potential decrease in the systemic exposure (i.e., serum concentration) of the affected drug, which could lead to a reduction or complete loss of its therapeutic efficacy.[2]
In vitro and clinical studies have identified several medications whose absorption can be significantly reduced by concurrent administration with patiromer. These include:
Conversely, studies have also shown that for some other commonly used oral medications, there is no clinically significant interaction, and no separation of dosing is required. This group includes drugs such as amoxicillin, lisinopril, losartan, furosemide, and metoprolol.[23]
The primary and most effective strategy for managing these potential interactions is temporal separation of drug administration. The universal recommendation is to administer other oral medications at least 3 hours before or 3 hours after taking patiromer.[2] This three-hour window is designed to allow most other oral drugs sufficient time to be absorbed from the upper GI tract before patiromer, which acts primarily in the colon, can interfere with their absorption.
This recommendation has undergone a significant regulatory evolution that has greatly impacted the drug's practicality in clinical use. Upon its initial FDA approval in 2015, patiromer carried a highly restrictive "boxed warning"—the FDA's most serious warning—mandating a 6-hour separation interval between its administration and that of any other oral drug.[8] This created substantial logistical challenges for patients, particularly the elderly and those with CKD or HF, who are often on complex, multi-drug regimens with doses scheduled throughout the day. Adhering to a 6-hour separation window was often impractical and posed a significant barrier to its use.
However, based on further data and analysis, the FDA approved a supplemental New Drug Application on November 27, 2016, which led to the removal of the boxed warning and a revision of the dosing separation advice.[41] The recommendation was changed to the current, less stringent 3-hour interval, and it was clarified that this separation applies specifically to drugs known to interact, not universally to all oral medications. This data-driven de-escalation of the perceived interaction risk was a critical step in the drug's post-marketing history. It transformed patiromer from a clinically effective but logistically difficult medication into a much more practical and manageable therapeutic option, thereby improving its utility and likely enhancing patient adherence to both patiromer and their other essential medications.
The dosing of patiromer must be individualized and adjusted based on regular monitoring of serum potassium levels to achieve and maintain the desired target range.
Proper preparation and administration are crucial for the efficacy and tolerability of patiromer. Patients should be counseled on the following steps:
To ensure the safe and effective use of patiromer, regular laboratory monitoring is mandatory.
Patiromer, developed by Relypsa, Inc. (later acquired by Vifor Pharma), underwent a structured regulatory review by the U.S. Food and Drug Administration (FDA).[3] The key milestones in its U.S. approval history are as follows:
Patiromer also underwent a thorough review process for authorization in the European Union.
The EMA's European Public Assessment Report (EPAR) concluded that the benefits of Veltassa in effectively reducing and controlling serum potassium levels outweighed its risks.[39] The risks were identified primarily as moderate gastrointestinal side effects and the potential for hypomagnesemia, both of which were deemed manageable through clinical monitoring and patient counseling.[39]
Following its approval in the U.S. and EU, patiromer has been authorized for use in numerous other countries worldwide.[12] However, its adoption and reimbursement have faced challenges related to pharmacoeconomics. As a novel, branded medication, its cost is substantially higher than that of older, generic potassium binders. This has led to scrutiny from healthcare payers and reimbursement bodies.
A notable example comes from a review conducted by PharmaCare in British Columbia, Canada. The review acknowledged patiromer's effectiveness in reducing serum potassium levels but noted the lack of direct comparative studies against other agents.[22] A pharmacoeconomic analysis conducted as part of this review suggested that a price reduction of at least 85% would be necessary for patiromer to be considered cost-effective within that healthcare system. After negotiations between the pan-Canadian Pharmaceutical Alliance (pCPA) and the manufacturer concluded without an agreement on price, the drug was designated as a "Non-Benefit" in that province.[22] This case highlights the significant tension that can exist between demonstrated clinical innovation and the budgetary constraints of public and private healthcare systems, which can ultimately impact patient access to new therapies.
Patiromer (Veltassa) is a significant and innovative addition to the therapeutic armamentarium for managing chronic hyperkalemia. As a non-absorbed, sodium-free, spherical polymer, its design represents a considerable pharmacotechnical advancement over older cation exchange resins, offering a more favorable safety and tolerability profile that is suitable for long-term use. Its mechanism of action, localized entirely within the gastrointestinal tract, provides a predictable pharmacokinetic and safety profile, which is particularly beneficial for the complex, polymedicated patients with chronic kidney disease and heart failure who constitute its primary user base.
The robust clinical evidence from the OPAL-HK and AMETHYST-DN trials has unequivocally demonstrated its efficacy in both lowering serum potassium to normal levels and, crucially, maintaining normokalemia over the long term. The most profound clinical contribution of patiromer is its role as an enabling therapy, allowing for the optimization of life-saving RAAS inhibitor therapy in high-risk cardiorenal patients by mitigating the dose-limiting side effect of hyperkalemia.
While its safety profile is generally benign, characterized primarily by manageable gastrointestinal side effects, clinicians must remain vigilant in monitoring for electrolyte disturbances, particularly hypomagnesemia. Furthermore, the potential for drug-drug interactions necessitates careful management through the temporal separation of administration. The evolution of its regulatory labeling regarding these interactions reflects a maturing understanding of its real-world risk profile and has substantially improved its clinical practicality. As with many novel therapies, pharmacoeconomic considerations remain a key factor in its accessibility, but its established clinical value in managing a potentially life-threatening condition solidifies its important place in modern cardiorenal pharmacotherapy.
Published at: August 15, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.