Furthering Managed Care Through Advances in IgA Nephropathy Therapy

Publication
Article
Supplements And Featured PublicationsAdvances in the Treatment of IgA Nephropathy

Introduction

Immunoglobulin A nephropathy (IgAN), also known as Berger disease, is an autoimmune condition and the most common type of primary glomerulonephritis worldwide.1,2 IgAN is charac­terized by the accumulation of IgA complexes in the kidney’s glomeruli, which results in pro­gressive damage that can lead to chronic kidney disease (CKD).3 The deposition of these antibodies triggers chronic renal inflammation that mani­fests as hematuria and proteinuria.3 The ongoing glomerular damage disrupts the filtration barrier; over time, scarring of the nephrons occurs, culminating in end-stage renal disease (ESRD).3 Alarmingly, 40% to 53% of patients progress to ESRD within two decades of diagnosis.4,5 This severe progression necessitates interventions such as dialysis or kidney transplantation; either of these interventions significantly impacts the patient’s quality of life (QOL) and imposes sub­stantial economic burdens.3

The landscape of IgAN treatment is rapidly evolving. As of September 2024, there are ongoing or recently completed phase 3 trials to investigate the safety and efficacy of at least 11 pharmaceutical agents to manage IgAN.6-16

This supplement reviews the clinical and economic impact of IgAN, the treatment land­scape including guideline recommendations and investigational agents, and managed care consid­erations for the coming months, when results of several phase 3 trials of investigational agents are anticipated.

Disease Overview

In the United States, the incidence of IgAN is approximately 1.29 cases per 100,000 individuals, amounting to over 4,000 diagnoses annually.5 Genetic factors play a crucial role in the patho­genesis of IgAN.17,18 The serum level of galactose deficient IgA1 (Gd-IgA1) is an inherited trait across various racial and ethnic groups.3 In most families, IgAN is transmitted in an autosomal dominant manner with incom­plete penetrance.3 Prevalence varies considerably among ethnic groups; it is highest among those of Asian descent, lower among Caucasians, and lowest among persons of sub-Saharan African heredity.19 The risk of developing IgAN is higher among males; in North America, the prevalence is twice as high among men as it is among women.19 Confirmed diagnosis of IgAN requires a kidney biopsy; therefore, the true prevalence and inci­dence may be underestimated due to underscreen­ing in subclinical presentations.

PATHOPHYSIOLOGY

In IgAN, the core mechanism of disease involves the formation and deposition of IgA complexes in the kidneys, triggering an inflammatory immune response that leads to tissue damage and clinical sequelae such as hematuria and proteinuria.20 With sustained glomerular injury, chronic inflam­mation persists and leads to progressive glomeruli necrosis and kidney failure.3

The pathophysiology of IgAN has been characterized as a multi-hit hypothesis involving four steps that combine to result in irreversible damage to the kidney.20-22 The first hit consists of increased circulating levels of Gd-IgA1, which is likely produced due to genetic factors.21 The second involves the production of glycan-specific IgG or IgA1 autoantibodies; this is usually preceded by an immune-mediated event such as a viral or bacterial infec­tion.3,22 Respiratory or intestinal infections may instigate the onset of the disease by triggering the production of these autoantibodies.3 The third hit occurs when immune complexes form in the circulation and reach high serum levels.22 When the immune complexes cross the glomerular filtration barrier to reach the mesangium, the complement cascade is activated and inflammatory mediators are produced, which constitutes the fourth hit.21,22 This multi­modal attack results in the pathophysiologic environment for kidney injury but offers the potential for researchers and drug manufacturers to intervene at multiple stages in the inflammatory process.

Disease Progression and Clinical Impact

The progression and clinical impact of IgAN can vary sig­nificantly among patients, ranging from a benign, asymp­tomatic condition to an aggressive form of the disease.17,20 In its mildest form, IgAN may be asymptomatic and can even resolve on its own.3 Clinically, it most frequently presents as microscopic hematuria and moderate protein­uria with preserved kidney function, usually following an infection.3 Microscopic hematuria with or without con­current proteinuria may be detected during routine health screenings and urinalysis.17,19 Episodic, visible hematuria that manifests as pink or cola-colored urine is the hallmark of the disease.17,19 Less commonly, patients might experience nephrotic-range proteinuria that can lead to leg, foot, and ankle edema and foamy urine.3,17

Within 20 years of diagnosis, up to half of patients with IgAN will progress to ESRD.5 Signs and symptoms of ESRD include hypertension, edema, fatigue, reduced urine output, weight loss, sleep disturbances, difficulty con­centrating, darkened skin and muscle cramps.3 Advanced stages of IgAN can lead to complications that include hypertension, chronic or acute renal disease, cardiovas­cular disease, nephrotic syndrome and Henoch-Schönlein purpura.3 Similar to other chronic nephritic diseases, IgAN is associated with an increased risk of mortality.3 In a 2018 study, researchers identified a 10.1-year reduction in life expectancy relative to predicted age of death at the time of kidney biopsy in patients with IgAN.23 Not surprisingly, 82.5% of the deaths occurred after progression to ESRD, underscoring the high burden of mortality associated with disease progression.23

Economic Impact

Although economic data on IgAN are limited, the man­agement of ESRD remains a significant cost burden to the U.S. healthcare system, highlighting the need for treat­ments that slow kidney disease progression and maintain health-related QOL (HRQOL).24-26 Approximately 15% of the U.S. population, or about 51 million people, are living with CKD.27,28 Healthcare expenditures for patients with CKD are high compared to those for their matched non-CKD cohorts, with significant increases in costs associ­ated with each advance in the stage of the disease.24,26 A large-scale retrospective analysis of health plan electronic medical records from 2007 to 2012 in the U.S. demon­strated that for patients with CKD who were younger than 65 years, the average annualized cost per patient increased at least 57% with each CKD stage. For patients 65 years or older, costs increased by at least 68% when disease progressed to ESRD or stages 4 or 5 CKD but by no more than 45% when disease progressed to stage 3a or 3b CKD. Inpatient admissions accounted for the bulk of increased costs in ESRD and stages 3b or 4 to 5 CKD; at least 80% of the cost increase was propelled by rising inpatient costs.26

In addition to inpatient expenses, healthcare costs in CKD are inflated by spending for dialysis treatment. In 2019, Medicare costs for CKD were estimated to exceed $87 billion with more than $37 billion spent on treating ESRD.29 Adjusted for inflation to July 2024 costs, this rep­resents Medicare costs of more than $108 billion for CKD and $46 billion for ESRD.30 For patients needing dialysis, the average annual Medicare expenditure is around $85,000 for outpatient-based hemodialysis and $69,000 for home-based peritoneal dialysis.24

Patients with IgAN experience a substantial symptom burden with pain and fatigue being the most commonly reported issues.3 In a systematic literature review of IgAN that evaluated eight studies on HRQOL, proteinuria was linked to lower HRQOL and higher rates of mental depression.5 Additionally, patients with glomerulonephri­tis reported poorer mental health compared with healthy individuals.5

These findings emphasize the need for early detection and treatment to minimize CKD progression, reduce healthcare costs and improve QOL for patients with IgAN.

Treatment Landscape

The primary aim of IgAN treatment is to prevent or delay progression to ESRD and preserve kidney func­tion.17,31 In its 2021 clinical practice guideline for the management of glomerular diseases, Kidney Disease: Improving Global Outcomes (KDIGO) issued guidance for the management of IgAN absent any FDA-approved treatments.31 Recent advancements in understanding the pathogenesis of IgAN as an immune-mediated disease and the acceptance of surrogate outcomes for accelerated drug approval have led to a rapid increase in clinical trials exploring new therapeutic strategies that target specific disease pathways.22,32,33 In December 2021 and February 2023, respectively, the FDA granted accelerated approval to budesonide (Tarpeyo; Calliditas Therapeutics) and sparsentan (Filspari; Travere Therapeutics) for reduction of proteinuria in adults with IgAN to reduce disease progression.34,35 In December 2023, the FDA granted full approval to budesonide to reduce loss of kidney function in all patients with IgAN who are at risk for disease progression; this marked the first drug approval for IgAN.36 In August 2024, the FDA granted accelerated approval to iptacopan (Fabhalta; Novartis) for reduction of proteinuria in adults with primary IgAN at risk of rapid disease progression.37,38 In September 2024, the FDA granted full approval to sparsentan to slow kidney function decline in adults with primary IgAN who are at risk of disease progression.35 As of September 2024, phase 3 trials investigating the safety and efficacy of at least 11 phar­maceutical agents to treat IgAN are ongoing or have been completed. In the coming months, results of many of these trials and regulatory decisions will transform the current treatment landscape for IgAN.

TREATMENT GUIDELINES

The KDIGO 2021 guidelines for IgAN include recom­mendations for diagnosis, prognosis and treatment.31 KDIGO emphasizes that kidney biopsy provides the only conclusive evidence to support a diagnosis of IgAN, as no validated diagnostic serum or urine biomarkers exist for the disease apart from estimated glomerular filtration rate (eGFR) and proteinuria (Table 1).31 Patients with IgAN should be assessed for secondary causes, and their disease should be stratified according to mesangial and endocap­illary hypercellularity, segmental sclerosis, interstitial fibrosis/tubular atrophy and crescents (MEST-C) score using what is known as the revised Oxford classification.31 The work group highlights that the International IgAN Prediction Tool should be used to quantify risk of disease progression and inform shared decision-making with patients.31

Guideline-Directed Treatment

With no FDA-approved treatments at the time of its writ­ing, the 2021 KDIGO guidelines underscore strategies not based upon immunosuppression.31 Aiming to slow disease progression, this supportive care is centered upon lifestyle modification (e.g., exercise, smoking cessation, weight loss, reduction of sodium in the diet), blood pressure control and renin-angiotensin system (RAS) inhibition.31

Cardiovascular risk should be addressed, and all patients should have their blood pressure managed, with the KDIGO work group suggesting a target systolic blood pressure of less than 120 mm Hg in most patients.31 RAS inhibition should be provided to all patients with IgAN who have proteinuria exceeding 0.5 g/day regard­less of hypertension status; therapy with either an angiotensin-converting enzyme inhibitor or angiotensin II receptor blocker should be used.31

For patients who remain at high risk of progressive CKD (proteinuria > 0.75-1 g/day) despite at least 90 days of optimized supportive care, the work group recommends offering participation in a clinical trial and consideration of immunosuppressive therapy, including a six-month course of glucocorticoids.31 However, glucocorticoids should be given with caution or, in certain situations (e.g., diabetes, obesity, severe osteoporosis), avoided entirely, as they do not impart a proven clinical benefit in IgAN.31

APPROVED AND INVESTIGATIONAL AGENTS IN PHASE 3 TRIALS

As of September 2024, phase 3 trials investigating the safety and efficacy of at least 11 pharmaceutical agents to treat IgAN are ongoing or have been completed (Table 2).6-16,22,37,39-44 Investigated strategies include use of a targeted-release steroid, anticomplement therapies, modulators of lym­phocyte activity and anti-endothelin or anti-aldosterone antagonists.

Corticosteroid

The targeted-release formulation of budesonide (TRF-budesonide) has been developed to deliver the drug directly to the gut mucosal level and to target gut mucosal inflammation and synthesis of Gd-IgA1.6 In the phase 3 NefIgArd trial (NCT03643965), the safety and efficacy of oral TRF-budesonide, 16 mg/day, was compared with those of placebo over a nine-month treatment period in patients with IgAN who experienced persistent proteinuria despite optimized supportive care.6 Patients were then followed for a 15-month observational period off study drug.6 At two-year follow up, the results showed a treatment benefit with TRF-budesonide over placebo in terms of time-weighted average of GFR, with a GFR change of -2.47 mL/min/1.73 m2 (95% CI, -3.88 to -1.02 mL/min/1.73 m2) in the TRF-budesonide group compared with -7.52 mL/min/1.73 m2 (95% CI, -8.83 to -6.18 mL/min/1.73 m2) in the placebo group, indicating approximately 50% less deterioration of kidney function in the treatment group.6 TRF-budesonide was granted accelerated approval by the FDA in December 2021 to reduce proteinuria in adults with IgAN and full approval in December 2023 to reduce the loss of kidney function in IgAN.34,36,39

ANTI-ENDOTHELIN ANTAGONISTS

Among anti-endothelin agonsists, both a dual endothelin receptor type A and angiotensin II receptor type 1 antag­onist (DEARA) and an endothelin receptor type A (ETAR) antagonist are being studied for IgAN treatment.

DEARA

Sparsentan is a nonimmunosuppressive, single-molecule DEARA with high selectivity for the ETAR and angiotensin II receptor type 1.7 The phase 3 PROTECT study (NCT03762850) was a multinational, double-blind, randomized, active-controlled study in which the safety and efficacy of sparsentan versus irbesartan were evaluated in patients with IgAN.7 The primary end point was proteinuria change at 36 weeks, with patients randomly assigned to receive a target dose of sparsentan, 400 mg, orally once daily or irbesartan, 300 mg, once daily.7 At the 36-week interim analysis, treat­ment with sparsentan resulted in a significant reduction from baseline in proteinuria (-49.8%) versus irbesartan (-15.1%), a relative reduction of 41% (least-squares mean ratio, 0.59; 95% CI, 0.51-0.69; p-value < 0.0001), which was maintained through 110 weeks of follow-up.7,45 In September 2024, the FDA granted full approval to sparsentan to slow kidney function decline in adults with primary IgAN who are at risk of disease progression.35

ETAR ANTAGONIST

Atrasentan (Novartis), an oral ETAR antagonist, is being evaluated in the phase 3 ALIGN study (NCT04573478) in patients with IgAN.11,46 The global, randomized, multi­center, double-blind, placebo-controlled trial enrolled approximately 320 patients to evaluate the efficacy and safety of atrasentan versus placebo in patients with IgAN.11 Participants are being randomly assigned to receive once-daily atrasentan, 0.75 mg, or placebo orally for 2.5 years as they continue to receive a maximally tolerated dose of a RAS inhibitor.11,46 The primary end point is change in proteinuria from baseline to 136 weeks, and topline results are expected in the first quarter of 2026.46 In October 2023, Novartis announced that the study met its primary end point at the 36-week interim analysis and plans to review those results with the FDA to allow for a potential regulatory submission for acceler­ated approval.46

Anti-Aldosterone Antagonist

Finerenone (Kerendia; Bayer HealthCare Pharmaceuticals, Inc.) is a nonsteroidal, selective miner­alocorticoid receptor antagonist that reduces the risks of CKD progression and cardiovascular events in patients with CKD and type 2 diabetes.47 The large, randomized, double-blind, placebo-controlled, phase 3 FIND-CKD trial (NCT05047263) is currently underway to evaluate the effect of finerenone in the nondiabetic CKD patient population including individuals with IgAN.12 An esti­mated 1,584 participants will be randomly assigned to take finerenone, 10 mg or 20 mg, orally once daily or pla­cebo for 50 months.12 Mean rate of change in eGFR from baseline to month 32 will be measured as the primary outcome.12

Anticomplement Therapies

The complement system is another target for IgAN therapy. Agents targeting the alternative pathway and complement pathway are being investigated.

ALTERNATIVE PATHWAY INHIBITION FACTOR B INHIBITORS

Iptacopan is a proximal complement inhibitor that specifically binds to factor B and inhibits the alterna­tive complement pathway, a potential target for IgAN pathophysiology.43 The ongoing phase 3 APPLAUSE-IgAN trial (NCT04578834) evaluates the efficacy and safety of iptacopan in managing IgAN.8,43 Patients with IgAN have been randomly assigned to iptacopan, 200 mg, or placebo twice daily orally during a 24-month treatment period; the primary objectives are to establish the superiority of iptacopan over placebo in reducing proteinuria at 9 months and to demonstrate a slowing of the rate of eGFR decline over 24 months.8,43 In October 2023, Novartis announced positive results from the prespecified interim analysis, indicating that iptacopan demonstrated supe­riority versus placebo in proteinuria reduction at nine months.48 In August 2024, the FDA granted accelerated approval to iptacopan for reduction of proteinuria in adults with primary IgAN at risk of rapid disease progres­sion.37,38 Study completion is expected in 2025.38

RO7434656 (IONIS FB-LRx; Roche) is another factor B complement inhibitor and is being studied in the phase 3 IMAGINATION trial (NCT05797610).10,22 The trial cur­rently is in the recruiting phase. In all, 428 eligible study participants will be enrolled and randomly assigned 1:1 to receive RO7434656 or placebo subcutaneously (SC) on days 1, 15 and 29 and every four weeks thereafter for 105 weeks.10 The primary end point is change from baseline in 24-hour urine protein-to-creatinine ratio (UPCR) at 37 weeks.10

COMPLEMENT C5 INHIBITOR

Ravulizumab (Ultomiris; Alexion Pharmaceuticals) is a humanized monoclonal antibody that inhibits com­plement C5; it is approved in the U.S. for the treatment of paroxysmal nocturnal hemoglobinuria, atypical hemolytic uremic syndrome, anti-acetylcholine receptor antibody-positive generalized myasthenia gravis, and AQP4 antibody-positive neuromyelitis optica spectrum disorder.49 Investigators are currently recruiting for the phase 3 ICAN study (NCT06291376), a randomized, double-blind, placebo-controlled study to evaluate the efficacy and safety of ravulizumab in patients with IgAN.9 Approximately 450 patients will be enrolled and randomly assigned to receive a weight-based intravenous infusion of either ravulizumab or placebo on days 1 and 15 and then every eight weeks thereafter. An interim analysis is planned for week 34 to evaluate change in proteinuria; the final analysis will be conducted at week 106 to evalu­ate eGFR.9

Lymphocyte Activity Modulators

In the treatment of IgAN, the modulation of lymphocyte activity is also being evaluated. Strategies include anti-proliferation-inducing ligand (APRIL) antibodies and dual anti-cytokine B-cell activating factor (BAFF) and APRIL antibodies.

APRIL Antibodies

Sibeprenlimab (VIS649; Otsuka Pharmaceutical) is an investigational humanized immunoglobulin G2 monoclo­nal antibody that reduces the production of Gd-IgA1 by binding to APRIL.50 In the ongoing phase 3 VISIONARY study (NCT05248646), the effects of sibeprenlimab, 400 mg, given SC every four weeks versus placebo on pro­teinuria and eGFR are being evaluated in 530 enrolled participants with IgAN.13 The primary outcome measure is 24-hour UPCR at nine months, with study completion estimated in 2026.13

Zigakibart (BION-1301; Novartis) is another novel humanized anti-APRIL monoclonal antibody that is in phase 3 development to manage IgAN.14,51 The BEYOND study (NCT05852938) is a global, randomized, double-blind, placebo-controlled trial in which investiga­tors will randomly assign approximately 272 patients with biopsy-proven IgAN to receive zigakibart, 600 mg, or a matched placebo SC every two weeks for 104 weeks. The primary end point is the change in proteinuria at 40 weeks.14

Dual Anti-Cytokine BAFF and APRIL Antibodies

Atacicept (Vera Therapeutics) is an investigational dual inhibitor of BAFF and APRIL.41,44 Its safety and efficacy in IgAN treatment is being evaluated in the ORIGIN 3 phase 3 clinical trial (NCT04716231), a multinational, random­ized, double-blind, placebo-controlled study that is currently recruiting.15 An estimated 376 study participants will be randomly assigned 1:1 to receive atacicept, 150 mg, once weekly by SC injection or placebo SC injections once weekly for 104 weeks, followed by a 52-week open-label extension.15 The primary end point is the change in proteinuria at week 36.15

Another dual BAFF-APRIL inhibitor is telitacicept (RemeGen Co.), which is also being investigated for efficacy and safety in IgAN.16 Investigators involved with a phase 3 trial (NCT05799287) currently are recruiting patients in China.16 An estimated 308 patients will be ran­domly assigned 1:1 either to telitacicept, 240 mg, SC given once weekly (phase A) for 39 doses and then every two weeks (phase B) for 32 doses or to placebo.16 The primary end points are the change in proteinuria from baseline at week 39 and the annualized eGFR slope at week 104.16

ADDITIONAL STRATEGIES

A phase 3 clinical trial (NCT02682407) for narsoplimab (OMS721; Omeros), a humanized monoclonal antibody targeting the MASP-2 pathway, was terminated for insufficient antiproteinuric effect compared with pla­cebo.52,53 The phase 3 BRILLIANT- SC trial (NCT02052219) evaluating blisibimod (Anthera), a BAFF antagonist, was withdrawn in 2021.54,55

Originally implemented for the treatment of type 2 diabetes, use of SGLT2 inhibitors has demonstrated significant improvements in cardiovascular and renal out­comes for both diabetic and nondiabetic CKD patients, including those with IgAN.22 A meta-analysis of 13 randomized controlled trials pooled the effects of SGLT2 inhibitors, demonstrating that SGLT2 inhibitors reduced the relative risk of kidney disease progression in patients with IgAN by 49% (95% CI, 0.32%-0.74%).56 In studies involving large patient populations with IgAN, SGLT2 inhibitors are being investigated for use in kidney disease; these studies include the DAPA-CKD (NCT03036150) and EMPA-KIDNEY (NCT03594110) trials.57,58

Additional strategies being investigated in phase 2 trials include MASP-2 inhibition, neutralization of the CD40 ligand, inhibition of factor D, suppression of both the alternative and terminal pathways of the complement system and neutralization of both APRIL and BLyS/BAFF.22,59-63

Managed Care Considerations

Managing IgAN requires comprehensive strategies to increase disease awareness, address its underdiagnosis and improve patient outcomes (Table 3).3,64-70 If left undi­agnosed and untreated, IgAN carries a significant mortal­ity risk; further, it is closely linked to the development of ESRD.19 Early diagnosis and treatment intervention may delay disease progression, reduce overall healthcare use and extend the lifespan of patients.23,25 Care management programs that focus on the early detection and treatment of IgAN may reduce healthcare expenditures on renal replacement therapies and improve patient outcomes by slowing disease progression.64

Community-based screening programs that are integrated into population health interventions can help with early identification of high-risk patients and allow for timely medical interventions to delay CKD progres­sion.64,65 A screening tool as simple as urinalysis may be used to detect urinary markers suggestive of glomerular injury. For instance, the National Kidney Foundation Kidney Early Evaluation Program uses dipstick urinalysis for hematuria; this method has been shown to reduce costs for mass screenings.66,67 The use of novel biomarkers (e.g., Gd-IgA1, Gd-IgA1 antibodies and Gd-IgA1 immune complexes) is being evaluated to possibly enhance clinical screening for IgAN.68,69

Early risk stratification and intervention including timely access to antihypertensives and supportive care for patients with CKD has demonstrated cost savings and improved patient outcomes in renal disease. While not limited to IgAN, a pilot patient-centered medical home program was implemented by CareFirst Blue Cross Blue Shield of Maryland from 2015 to 2017 to identify high-risk patients and enroll them in kidney-related care plans that provided kidney testing, patient education on lifestyle modifications, disease awareness, nephrologist referral and medication management.65 The program demon­strated a per-member, per-month cost savings in the first year due to reduced hospital admissions for patients having stage 3 or 5 CKD; authors anticipated future improvements in rates of patients progressing to dialysis over a longer period of time.65

Managed care professionals can play a crucial role in medication management of kidney disease. Polypharmacy is common among CKD and dialysis patients, who may use up to 10 to 12 medications on average; these patients are at increased risk for medication-related side-effects and drug interactions.70 In addition to optimizing phar­macotherapy, managed care professionals can support patient education initiatives to promote lifestyle modifi­cations.3 Provider knowledge of the safety and efficacy of available treatments is critical to the successful man­agement of renal disease. Timely incorporation of novel therapeutics ensures early patient access to treatment and promotes improved patient outcomes.3

Conclusions

IgAN is the most common cause of primary glomerular disease. It is associated with significant morbidity and mortality with increased risk of progression to ESRD in patients who are left untreated. Significant advances in understanding IgAN as an immune-mediated dis­ease have led to the development of targeted therapies currently being evaluated in clinical trials, potentially expanding therapeutic options for treating IgAN. Armed with the knowledge of investigational therapies and pipe­line drugs, managed care professionals can reduce overall healthcare expenditures by addressing gaps in treatment diagnosis, implementing early disease management and improving patient outcomes in the setting of IgAN.


REFERENCES

1. Pattrapornpisut P, Avila-Casado C, Reich HN. IgA nephropathy: core curricu­lum 2021. Am J Kidney Dis. 2021;78(3):429-441. doi:10.1053/j.ajkd.2021.01.024

2. Rout P, Limaiem F, Hashmi MF. IgA nephropathy (Berger disease). StatPearls. National Center for Biotechnology Information; 2024. https://www.ncbi.nlm.nih.gov/books/NBK538214/

3. Grabe DW. Preventing progression in IgA nephropathy: a managed care focus on emerging therapies. Am J Manag Care. 2023;29(suppl 3):S31-S43. doi:10.37765/ajmc.2023.89344

4. Tomino Y. Diagnosis and treatment of patients with IgA nephropa­thy in Japan. Kidney Res Clin Pract. 2016;35(4):197-203. doi:10.1016/j.krcp.2016.09.001

5. Kwon CS, Daniele P, Forsythe A, Ngai C. A systematic literature review of the epidemiology, health-related quality of life impact, and economic burden of immunoglobulin A nephropathy. J Health Econ Outcomes Res. 2021;8(2):36-45. doi:10.36469/001c.26129

6. Lafayette R, Kristensen J, Stone A, et al; NefIgArd trial investigators. Efficacy and safety of a targeted-release formulation of budesonide in patients with primary IgA nephropathy (NefIgArd): 2-year results from a randomised phase 3 trial. Lancet. 2023;402(10405):859-870. doi:10.1016/S0140-6736(23)01554-4

7. Rovin BH, Barratt J, Heerspink HJL, et al; DUPRO steering committee and PROTECT Investigators. Efficacy and safety of sparsentan versus irbesartan in patients with IgA nephropathy (PROTECT): 2-Year results from a randomised, active-controlled, phase 3 trial. Lancet. 2023;402(10417):2077-2090. doi:10.1016/S0140-6736(23)02302-4

8. Study of efficacy and safety of LNP023 in primary IgA nephropathy patients (APPLAUSE-IgAN). ClinicalTrials.gov. Updated July 15, 2024. Accessed August 2, 2024. https://clinicaltrials.gov/study/NCT04578834

9, Study of ravulizumab in immunoglobulin A nephropathy (IgAN) (ICAN). ClinicalTrials.gov. Updated July 19, 2024. Accessed July 22, 2024. https://clinicaltrials.gov/study/NCT06291376

10. A study to evaluate the efficacy and safety of RO7434656 in participants with primary immunoglobulin A (IgA) nephropathy at high risk of progression (IMAGINATION). ClinicalTrials.gov. Updated July 8, 2024. Accessed July 9, 2024. https://clinicaltrials.gov/study/NCT05797610

11. Atrasentan in patients with IgA nephropathy (ALIGN). ClinicalTrials.gov. Updated May 23, 2024. Accessed July 9, 2024. https://clinicaltrials.gov/study/NCT04573478

12. A trial to learn how well finerenone works and how safe it is in adult partic­ipants with non-diabetic chronic kidney disease (FIND-CKD). ClinicalTrials.gov. Updated July 1, 2024. Accessed July 9, 2024. https://clinicaltrials.gov/study/NCT05047263

13. Visionary study: phase 3 trial of sibeprenlimab in immunoglobulin A nephropathy (IgAN). ClinicalTrials.gov. Updated March 26, 2024. Accessed July 22, 2024. https://clinicaltrials.gov/study/NCT05248646

14. A study of BION-1301 in adults with IgA nephropathy. ClinicalTrials.gov. Updated May 20, 2024. Accessed July 22, 2024. https://clinicaltrials.gov/study/NCT05852938

15. Atacicept in subjects with IgA nephropathy (ORIGIN 3). ClinicalTrials.gov. Updated November 29, 2023. Accessed July 22, 2024. https://clinicaltrials.gov/study/NCT04716231

16. A study of telitacicept in patients with primary IgA nephropathy. ClinicalTrials.gov. Updated September 6, 2023. Accessed July 22, 2024. https://clinicaltrials.gov/study/NCT05799287

17. IgA nephropathy. National Institute of Diabetes and Digestive and Kidney Diseases. Updated September 2022. Accessed July 10, 2024. https://www.niddk.nih.gov/health-information/kidney-disease/iga-nephropathy

18. Kiryluk K, Novak J. The genetics and immunobiology of IgA nephropathy. J Clin Invest. 2014;124(6):2325-2332. doi:10.1172/JCI74475

19. Rajasekaran A, Julian BA, Rizk DV. IgA nephropathy: an interesting autoimmune kidney disease. Am J Med Sci. 2021;361(2):176-194. doi:10.1016/j.amjms.2020.10.003

20. Wyatt RJ, Julian BA. IgA nephropathy. N Engl J Med. 2013;368(25):2402-2414. doi:10.1056/NEJMra1206793

21. Canetta PA, Kiryluk K, Appel GB. Glomerular diseases: emerging tests and therapies for IgA nephropathy. Clin J Am Soc Nephrol. 2014;9(3):617-625. doi:10.2215/CJN.07260713

22. Del Vecchio L, Allinovi M, Comolli S, Peiti S, Rimoldi C, Locatelli F. Drugs in development to treat IgA nephropathy. Drugs. 2024;84(5):503-525. doi:10.1007/s40265-024-02036-1

23. Hastings MC, Bursac Z, Julian BA, et al. Life expectancy for patients from the southeastern United States with IgA nephropathy. Kidney Int Rep. 2018;3(1):99-104. doi:10.1016/j.ekir.2017.08.008

24. Wang V, Vilme H, Maciejewski ML, Boulware LE. The economic burden of chronic kidney disease and end-stage renal disease. Semin Nephrol. 2016;36(4):319-330. doi:10.1016/j.semnephrol.2016.05.008

25. Nichols GA, Ustyugova A, Deruaz-Luyet A, OʼKeeffe, Rosetti M, Brodovicz KG. Health care costs by type of expenditure across eGFR stages among patients with and without diabetes, cardiovascular disease, and heart failure. J Am Soc Nephrol. 2020;31(7):1594-1601. doi:10.1681/ASN.2019121308

26. Golestaneh L, Alvarez PJ, Reaven NL, et al. All-cause costs increase exponentially with increased chronic kidney disease stage. Am J Manag Care. 2017;23(suppl 10):S163-S172.

27. Johansen KL, Chertow GM, Foley RN, et al. US Renal Data System 2020 annual data report: epidemiology of kidney disease in the United States. Am J Kidney Dis. 2021;77(4; suppl 1):A7-A8. doi:10.1053/j.ajkd.2021.01.002

28. U.S. and world population clock. United States Census Bureau. Accessed August 12, 2024. https://www.census.gov/popclock/

29. Chronic kidney disease. US Department of Health and Human Services. Centers for Disease Control and Prevention. Updated May 15, 2024. Accessed August 4, 2022. https://www.cdc.gov/kidney-disease/ckd-facts/

30. CPI inflation calculator. U.S. Bureau of Labor Statistics. Accessed July 17, 2024. https://www.bls.gov/data/inflation_calculator.htm

31. Kidney Disease: Improving Global Outcomes (KDIGO) Glomerular Diseases Work Group. KDIGO 2021 clinical practice guideline for the managementof glomerular diseases. Kidney Int. 2021;100(4S):S1-S276. doi:10.1016/j. kint.2021.05.021

32. Accelerated approval program. FDA. Updated February 2, 2024. Accessed July 9, 2024. https://www.fda.gov/drugs/nda-and-bla-approvals/ accelerated-approval-program

33. Ongoing | Non-malignant hematological, neurological, and other disorder indications accelerated approvals. FDA. Updated May 3, 2024. Accessed August 12, 2024. https://www.fda.gov/drugs/accelerated-approval-program/ ongoing-non-malignant-hematological-neurological-and-other-disorder-indi­cations-accelerated

34. FDA approves first drug to decrease urine protein in IgA nephropathy, a rare kidney disease. FDA. December 17, 2021. Accessed July 9, 2024. https:// www.fda.gov/drugs/fda-approves-first-drug-decrease-urine-protein-iga-ne­phropathy-rare-kidney-disease

35. Travere Therapeutics announces full FDA approval of Filspari (sparsentan). News release. Travere. September 5, 2024. Accessed September 17, 2024. https://ir.travere.com/news-releases/news-release-details/travere-therapeutics-announces-full-fda-approval-filsparir

36. NDA 215935/-003 (Tarpeyo supplement approval letter). FDA. December 20, 2023. Accessed August 2, 2024. https://www.accessdata.fda.gov/drug­satfda_docs/appletter/2023/215935Orig1s003ltr.pdf

37. Fabhalta. Prescribing information. Novartis; 2024. Accessed August 13, 2024. https://www.novartis.com/us-en/sites/novartis_us/files/fabhalta.pdf

38. Novartis receives FDA accelerated approval for Fabhalta (iptacopan), the first and only complement inhibitor for the reduction of proteinuria in primary IgA nephropathy (IgAN). News release. Novartis. August 8, 2024. Accessed August 13, 2024. https://www.novartis.com/news/media-releases/novartis-re­ceives-fda-accelerated-approval-fabhalta-iptacopan-first-and-only-comple­ment-inhibitor-reduction-proteinuria-primary-iga-nephropathy-igan

39. Tarpeyo. Prescribing information. Calliditas Therapeutics AB; 2024. Accessed July 9, 2024. https://www.tarpeyo.com/prescribinginformation.pdf

40. Filspari. Prescribing information. Travere Therapeutics, Inc.; 2023. Accessed July 9, 2024. https://filsparihcp.com/igan/filspari-prescribing-infor­mation.pdf

41. Vera Therapeutics initiates pivotal phase 3 (ORIGIN 3) clini­cal trial of atacicept for the treatment of IgA nephropathy (IgAN). News release. Vera Therapeutics. June 7, 2024. Accessed July 22, 2023. https://ir.veratx.com/news-releases/news-release-details/ vera-therapeutics-initiates-pivotal-phase-3-origin-3-clinical

42. Lv J, Liu L, Hao C, Li G, et al. Randomized phase 2 trial of telitacicept in patients with IgA nephropathy with persistent proteinuria. Kidney Int Rep. 2022;8(3):499-506. doi:10.1016/j.ekir.2022.12.014

43. Rizk DV, Rovin BH, Zhang H, et al. Targeting the alternative complement pathway with iptacopan to treat IgA nephropathy: design and rationale of the APPLAUSE-IgAN study. Kidney Int Rep. 2023;8(5):968-979. doi:10.1016/j. ekir.2023.01.041

44. Kaegi C, Steiner UC, Wuest B, Crowley C, Boyman O. Systematic review of safety and efficacy of atacicept in treating immune-mediated disorders. Front Immunol. 2020;11:433. doi:10.3389/fimmu.2020.00433

45. Heerspink HJL, Radhakrishnan J, Alpers CE, et al. Sparsentan in patients with IgA nephropathy: a prespecified interim analysis from a randomised, double-blind, active-controlled clinical trial. Lancet. 2023;401(10388):1584- 1594. doi:10.1016/S0140-6736(23)00569-X

46. Novartis investigational atrasentan phase III study demonstrates clinically meaningful and highly statistically significant proteinuria reduction in patients with IgA nephropathy (IgAN). News release. Novartis. October 30, 2023. Accessed July 22, 2024. https://www.novartis.com/news/media-releases/ novartis-investigational-atrasentan-phase-iii-study-demonstrates-clin­ically-meaningful-and-highly-statistically-significant-proteinuria-reduc­tion-patients-iga-nephropathy-igan

47. Kerendia. Prescribing information. Bayer HealthCare Pharmaceuticals Inc.; 2022. Accessed July 9, 2024. https://www.kerendia-us.com/pi

48. Novartis investigational iptacopan phase III study demonstrates clinically meaningful and highly statistically significant proteinuria reduction in patients with IgA nephropathy (IgAN). News release. Novartis. October 2, 2023. Accessed July 22, 2024. https://www.novartis.com/news/media-releases/ novartis-investigational-iptacopan-phase-iii-study-demonstrates-clinical­ly-meaningful-and-highly-statistically-significant-proteinuria-reduction-patients-iga-nephropathy-igan

49. Ultomiris. Prescribing information. Alexion Pharmaceuticals, Inc.; 2024. Accessed July 9, 2024. https://alexion.com/Documents/Ultomiris_USPI.pdf

50. Sibeprenlimab received U.S. FDA breakthrough therapy designation for the treatment of immunoglobulin A nephropathy (IgAN). News release. Novartis. February 16, 2024. Accessed July 22, 2024. https://www.otsuka-us. com/news/sibeprenlimab-receives-us-fda-breakthrough-therapy-designa­tion-treatment-immunoglobulin

51. Barrat J, Kim S, Agha I, et al. WCN23-1175 updated interim results of a phase 1/2 study of BION-1301 in patients with IgA nephropathy. Kidney Int Rep. 2023;8(suppl 3):S280-S281. doi: 10.1016/j.ekir.2023.02.632.

52. Study of the safety and efficacy of OMS721 in patients with immunoglobu­lin A (IgA) nephropathy. ClinicalTrials.gov. Updated March 15, 2024. Accessed July 22, 2024. https://clinicaltrials.gov/study/NCT03608033

53. Lafayette RA, Rovin BH, Reich HN, Tumlin JA, Floege J, Barratt J. Safety, tolerability and efficacy of narsoplimab, a novel MASP-2 inhibitor for the treat­ment of IgA nephropathy. Kidney Int Rep. 2020;5(11):2032-2041. doi:10.1016/j. ekir.2020.08.003

54. BRILLIANT -SC: a study of the efficacy and safety of blisibimod adminis­tration in subjects with IgA Nephropathy. ClinicalTrials.gov. Updated May 23, 2024. Accessed July 22, 2024. https://clinicaltrials.gov/study/NCT02052219

55. Lenert A, Niewold TB, Lenert P. Spotlight on blisibimod and its potential in the treatment of systemic lupus erythematosus: evidence to date. Drug Des Devel Ther. 2017;11:747-757. doi:10.2147/DDDT.S114552

56. Nuffield Department of Population Health Renal Studies Group; SGLT2 inhibitor Meta-Analysis Cardio-Renal Trialists’ Consortium. Impact of diabetes on the effects of sodium glucose co-transporter-2 inhibitors on kidney out­comes: collaborative meta-analysis of large placebo-controlled trials. Lancet. 2022;400(10365):1788-1801. doi:10.1016/S0140-6736(22)02074-8

57. Heerspink HJL, Stefánsson BV, Correa-Rotter R, et al; DAPA-CKD Trial Committees and Investigators. Dapagliflozin in patients with chronic kidney disease. N Engl J Med. 2020;383(15):1436-1446. doi:10.1056/NEJMoa2024816

58. The EMPA-KIDNEY Collaborative Group; Herrington WG, Staplin N, et al. Empagliflozin in patients with chronic kidney disease. N Engl J Med. 2023;388(2):117-127. doi:10.1056/NEJMoa2204233

59. Clinical study of CM338 in the treatment of immunoglobulin A nephrop­athy. ClinicalTrials.gov. Updated July 28, 2023. Accessed August 2, 2024. https://clinicaltrials.gov/study/NCT05775042

60. Safety and efficacy of AT-1501 in patients with IgA nephropathy (IgAN). ClinicalTrials.gov. Updated March 26, 2024. Accessed August 2, 2024. https:// clinicaltrials.gov/study/NCT05125068

61. Study of ALXN2050 in proliferative lupus nephritis (LN) or immunoglobulin A nephropathy (IgAN). ClinicalTrials.gov. Updated July 19, 2024. Accessed August 2, 2024. https://clinicaltrials.gov/study/NCT05097989

62. Efficacy, safety, pharmacokinetics, and pharmacodynamics of KP104 to treat glomerulonephritis. ClinicalTrials.gov. Updated August 14, 2023. Accessed August 2, 2024. https://clinicaltrials.gov/study/NCT05517980

63. An open-label study of povetacicept (ALPN-303) in autoimmune kidney diseases (RUBY-3). ClinicalTrials.gov. Updated July 19, 2024. Accessed August 2, 2024. https://clinicaltrials.gov/study/NCT05732402

64. Curtis S, Komenda P. Screening for chronic kidney disease: moving toward more sustainable health care. Curr Opin Nephrol Hypertens. 2020;29(3):333- 338. doi:10.1097/MNH.0000000000000597

65. Vassalotti JA, DeVinney R, Lukasik S, et al. CKD quality improvement intervention with PCMH integration: health plan results. Am J Manag Care. 2019;25(11):e326-e333.

66. Jurkovitz CT, Qiu Y, Wang C, Gilbertson DT, Brown WW. The Kidney Early Evaluation Program (KEEP): program design and demographic characteristics of the population. Am J Kidney Dis. 2008;51(4; suppl 2):S3-S12. doi:10.1053/j. ajkd.2007.12.022

67. Okubo R, Hoshi SL, Kimura T, et al. Cost-effectiveness of mass screening for dipstick hematuria in Japan [published correction appears in Clin Exp Nephrol. 2022;26(5):413-414. doi: 10.1007/s10157-022-02183-3]. Clin Exp Nephrol. 2022;26(5):398-412. doi:10.1007/s10157-021-02170-0

68. Gholaminejad A, Gheisari Y, Jalali S, Roointan A. Comprehensive analysis of IgA nephropathy expression profiles: identification of potential biomarkers and therapeutic agents. BMC Nephrol. 2021;22(1):137. doi:10.1186/ s12882-021-02356-4

69. Ohyama Y, Renfrow MB, Novak J, Takahashi K. Aberrantly glycosylated IgA1 in IgA nephropathy: what we know and what we don’t know. J Clin Med. 2021;10(16):3467. doi:10.3390/jcm10163467

70. Gedney N. The impact of medication cost on dialysis patients. Kidney360. 2021;2(6):922-923. doi:10.34067/KID.0002162021

© 2024 MJH Life Sciences

All rights reserved.