Commercial
Advanced Search

Lumasiran (Oxlumo™)
08.01.74

Policy

MEDICALLY NECESSARY


INITIAL APPROVAL


Lumasiran is considered medically necessary and, therefore, covered for the treatment of primary hyperoxaluria type 1 (PH1) to lower urinary oxalate levels in​ pediatric and adult individuals when ALL of the following criteria are met:

  1. Diagnosis of primary hyperoxaluria type 1 confirmed by identification of biallelic pathogenic variants in alanine:glyoxylate aminotransferase (AGT or AGXT) gene OR liver biopsy demonstrating AGT deficiency.

  2. Presence of one of the following, which are associated with clinical presentation of primary hyperoxaluria type 1:

    -- Elevated urine oxalate excretion (body surface area-normalized daily urine oxalate excretion output ≥0.7 mmol/1.73 m2)

    -- Elevated plasma oxalate concentration >20 μmol/L or >1.76 mg/L

    -- Urine oxalate excretion:creatinine ratio above age-specific upper limit of normal.

  3. Individual has not received a liver transplant.

  4. Prescribed by or in consultation with a nephrologist, urologist, geneticist, or any healthcare provider with expertise in treating primary hyperoxaluria type 1.

CONTINUATION OF TREATMENT

Continuation of lumasiran is considered medically necessary and, therefore, covered​ when ALL of the following requirements are met:

  1. Individual continues to meet the initial treatment criteria cited above.

  2. Documented evidence to support clinically meaningful response to therapy from pre-treatment baseline (e.g., decreased urinary oxalate concentrations, decreased urinary oxalate:creatinine ratio, decreased plasma oxalate concentrations, improvement, stabilization or slowed worsening of nephrocalcinosis, renal stone events, renal impairment or systemic calcinosis).

  3. Does not exceed United States Food and Drug Administration approved maintenance dose.

EXPERIMENTAL/INVESTIGATIONAL 


Lumasiran is considered experimental/investigational and, therefore, not covered when the above criteria are not met and for all other indications unless the indication is supported as an accepted off-label use, as defined in the Company medical policy on off-label coverage for prescription drugs and biologics.

REQUIRED DOCUMENTATION


The individual's medical record must reflect the medical necessity for the care provided. These medical records may include, but are not limited to: records from the professional provider's office, hospital, nursing home, home health agencies, therapies, and test reports.


The Company may conduct reviews and audits of services to our members, regardless of the participation status of the provider. All documentation is to be available to the Company upon request. Failure to produce the requested information may result in a denial for the drug.


Guidelines

BENEFIT APPLICATION


Subject to the terms and conditions of the applicable benefit contract, lumasiran (Oxlumo) is covered under the medical benefits of the Company's products when the medical necessity criteria listed in this medical policy are met.


U.S. Food and Drug Administration (FDA


On November 23, 2020, lumasiran (Oxlumo) was approved by the U.S. Food and Drug Administration (FDA) for the treatment of primary hyperoxaluria type 1 to lower urinary oxalate levels in pediatric and adult patients.


On October 6, 2022, the U.S. FDA approved a new label expansion to lower urinary oxalate and plasma oxalate levels in pediatric and adult patients. The approval was based on the results of the ILLUMINATE-C phase 3 trial in patients with severe renal impairment, including individuals on hemodialysis.


DOSAGE AND FREQUENCY

The recommended dose of lumasiran is weight-based and given as a subcutaneous injection. All maintenance doses begin 1 month after the last loading dose.


  • For individuals weighing less than 10 kg: Loading dose is 6 mg/kg once monthly for 3 doses followed by a maintenance dose of 3 mg/kg once monthly.

  • For individuals weighing 10 kg to less than 20 kg: Loading dose is 6 mg/kg once monthly for 3 doses followed by a maintenance dose of 6 mg/kg once every 3 months (quarterly).

  • For individuals weighing 20 kg and above: Loading dose is 3 mg/kg once monthly for 3 doses followed by a maintenance dose of 3 mg/kg once every 3 months (quarterly).

For individuals on hemodialysis, lumasiran is administered after hemodialysis if administered on dialysis days.


High-dose pyridoxine has been shown to be effective in reducing urinary oxalate levels in individuals with primary hyperoxaluria type 1, particularly those with homozygous p.Gly170Arg or p.Phe152lle variant. In the pivotal trials of lumasiran, background use of pyridoxine was allowed. About 50% and 61% of participants in the lumasiran-treated arm in ILLUMINATE-A and ILLUMINATE-B received background pyridoxine treatment.


Description

PRIMARY HYPEROXALURIA TYPE1 (PH1)


Primary hyperoxalurias (PHs) are rare inborn errors of glyoxylate metabolism characterized by the over-production of oxalate, which is poorly soluble and is deposited as calcium oxalate in various organs (including the bones, eyes, heart, and kidney).  Primary hyperoxaluria is caused by mutations in 1 of the 3 genes that encode enzymes involved in glyoxylate metabolism.  Primary hyperoxaluria type 1 (PH1; about 80 % of cases) is due to mutations of hepatic peroxisomal enzyme alanine-glyoxylate aminotransferase (AGT); it is the most common and severe type affecting an estimated 1 to 3 individuals per million in Europe and North America.  As oxalate is primarily excreted in the urine, the kidney is the prime target for oxalate deposition, which leads to end-stage renal disease (ESRD) in many cases.  Individuals with PH1 produce excessive oxalate, which can combine with calcium to cause kidney stones and deposits in the kidneys.  Individuals can experience progressive kidney damage, which can lead to kidney failure and the need for dialysis.  As kidney function worsens, oxalate can build up and damage other organs (Cochat and Rumsby, 2013; Niaudet, 2022).

 

Liebow and associates (2017) stated that PH1 arises from mutations in the enzyme AGT; and the resulting deficiency in this enzyme leads to abnormally high oxalate production resulting in calcium oxalate crystal formation and deposition in the kidney and many other organs, with systemic oxalosis and ESRD being common outcomes.  Although a small subset of individuals can manage the disease with vitamin B6 treatments, the only effective treatment for most is a combined liver-kidney transplant.  These researchers discussed the development of ALN-GO1, an investigational RNA interference (RNAi) therapeutic targeting glycolate oxidase, to deplete the substrate for oxalate synthesis.  Subcutaneous administration of ALN-GO1 resulted in potent, dose-dependent, and durable silencing of the mRNA encoding glycolate oxidase and increased serum glycolate concentrations in wild-type mice, rats, and non-human primates.  Furthermore, ALN-GO1 increased urinary glycolate concentrations in normal non-human primates and in a genetic mouse model of PH1.  Notably, ALN-GO1 reduced urinary oxalate concentration up to 50 % following a single-dose in the genetic mouse model of PH1, and up to 98 % after multiple doses in a rat model of hyperoxaluria.  The authors concluded that these findings showed the ability of ALN-GO1 to reduce oxalate production in pre-clinical models of PH1 across multiple species and provided a clear rationale for clinical trials with this compound.

 

McGregor and colleagues (2020) noted that by sequencing autozygous human populations, these researchers identified a healthy adult woman with lifelong complete knockout of HAO1 (expected about 1 in 30 million outbred people).  HAO1 (glycolate oxidase) silencing is the mechanism of lumasiran, an investigational RNAi therapeutic for PH1.  Lumasiran acts by lowering oxalate production.  Her plasma glycolate levels were 12 times, and urinary glycolate 6 times, the upper limit of normal observed in healthy reference individuals (n = 67).  Plasma metabolomics and lipidomics (1,871 biochemicals) revealed 18 markedly elevated biochemicals (greater than 5 standard deviations outliers versus n = 25 controls) suggesting additional HAO1 effects.  Comparison with lumasiran pre-clinical and clinical trial data suggested she had less than 2 % residual glycolate oxidase activity.  Cell line p.Leu333SerfsTer4 expression showed markedly reduced HAO1 protein levels and cellular protein mis-localization.  The authors concluded that in this individual, lifelong HAO1 knockout is safe and without clinical phenotype, de-risking a therapeutic approach and informing therapeutic mechanisms.  Unlocking evidence from the diversity of human genetic variation can facilitate drug development.

 

LUMASIRAN


Lumasiran, a HAO1-directed small interfering ribonucleic acid (siRNA), is available as the brand Oxlumo (Alnylam Pharmaceuticals, Inc.). Per the label for Oxlumo, lumasiran reduces levels of glycolate oxidase (GO) enzyme by targeting the hydroxyacid oxidase 1 (HAO1) messenger ribonucleic acid (mRNA) in hepatocytes through RNA interference. Decreased GO enzyme levels reduce the amount of available glyoxylate, a substrate for oxalate production. As the GO enzyme is upstream of the deficient alanine:glyoxylate aminotransferase (AGT) enzyme that causes PH1, the mechanism of action of lumasiran is independent of the underlying AGXT gene mutation. Oxlumo is not expected to be effective in primary hyperoxaluria type 2 (PH2) or type 3 (PH3) because its mechanism of action does not affect the metabolic pathways causing hyperoxaluria in PH2 and PH3 (Alnylam, 2022).

The most common adverse reaction (reported in 20% or more of individuals) is injection site reactions.

Lumasiran acts by lowering oxalate production.  It was examined in 2 studies in individuals with PH1: a randomized, placebo-controlled trial in individuals 6 years and older and an open-label study in individuals younger than 6 years.​



CLINICAL STUDIES


ILLUMINATE-A


ILLUMINATE-A was a randomized, double-blind trial comparing lumasiran and placebo in 39 individuals 6 years of age and older with PH1 and an eGFR ≥30 mL/min/1.73 m2 (ILLUMINATE-A; NCT03681184). Individuals received 3 loading doses of 3 mg/kg OXLUMO (N=26) or placebo (N=13) administered once monthly, followed by quarterly maintenance doses of 3 mg/kg OXLUMO or placebo.

The median age was 15 years (range 6 to 61 years), 67% were male, and 77% were White. At baseline, the median 24-hour urinary oxalate excretion corrected for body surface area (BSA) was 1.7 mmol/24 h/1.73 m2, the median plasma oxalate level was 13.1 µmol/L, 33% of individuals had eGFR ≥90 mL/min/1.73 m2, 49% had eGFR of 60 to <90 mL/min/1.73 m2, and 18% had eGFR 30 to <60 mL/min/1.73 m2, 56% were on pyridoxine, and 85% reported a history of symptomatic kidney stone events.

The primary endpoint was the percent reduction from baseline in 24-hour urinary oxalate excretion corrected for BSA averaged over Months 3 through 6. The LS mean percent change from baseline in 24­hour urinary oxalate in the OXLUMO group was -65% (95% CI: -71, -59) compared with -12% (95% CI: -20, -4) in the placebo group, resulting in a between-group LS mean difference of 53% (95% CI: 45, 62; p<0.0001). By Month 6, 52% (95% CI: 31, 72) of individuals treated with OXLUMO achieved a normal 24-hour urinary oxalate corrected for BSA (≤0.514 mmol/24 hr/1.73 m2) compared to 0% (95% CI: 0, 25) placebo-treated individuals (p=0.001).

 

ILLUMINATE-B


ILLUMINATE-B was a single-arm study in 18 individuals <6 years of age with PH1 and an eGFR >45 mL/min/1.73 m2 for individuals ≥12 months of age or a normal serum creatinine for individuals <12 months of age (ILLUMINATE-B; NCT03905694). Efficacy analyses included the first 16 individuals who received 6 months of treatment with OXLUMO. Dosing was based on body weight.

The median age was 47 months (range 4 to 74 months), 56% were female, and 88% were White. Three individuals were less than 10 kg, 11 were 10 kg to < 20 kg, and 2 were ≥ 20 kg. The median spot urinary oxalate:creatinine ratio at baseline was 0.47 mmol/mmol. The primary endpoint was the percent reduction from baseline in spot urinary oxalate:creatinine ratio averaged over Months 3 through 6. Individuals treated with OXLUMO achieved a reduction in spot urinary oxalate:creatinine ratio from baseline of 71% (95% CI: 65, 77).


SUMMARY


Primary hyperoxalurias are a group of rare genetic diseases. There are 3 subtypes each resulting in the overproduction of oxalate by the liver. Type 1 is the most common type, which accounts for approximately 80% of cases and occurs as a result of a genetic defect in the alanine:glyoxylate aminotransferase (AGXT) gene that encodes the enzyme alanine glyoxylate aminotransferase. Defect in the enzyme results in overproduction of oxalate, which leads to deposition of calcium oxalate crystals in the kidneys and urinary tract. The result is the formation of painful and recurrent nephrolithiasis (renal stones), nephrocalcinosis, and renal failure. Compromised renal function exacerbates the disease as the excess oxalate can no longer be effectively excreted, resulting in subsequent accumulation and crystallization in bones, eyes, skin, and heart, leading to severe illness and death. Lumasiran is a subcutaneously administered RNA interference (RNAi) therapeutic that silences the HAO1 gene, which encodes for a glycolate oxidase enzyme. By silencing the HAO1 gene, levels of glycolate oxidase are depleted, decreasing production of oxalate, the metabolite that directly contributes to the pathophysiology of primary hyperoxaluria type 1.

For individuals with primary hyperoxaluria type 1, evidence includes 1 phase 3 randomized controlled trial (RCT) (ILLUMINATE-A) in individuals 6 years and older and 2 single arm prospective studies (ILLUMINATE-B and ILLUMINATE-C). Relevant outcomes are symptoms, quality of life, disease-specific survival, change in disease status, treatment-related morbidity, and treatment-related mortality. In ILLUMINATE-A and ILLUMINATE-B, individuals with preserved renal function were enrolled (estimated glomerular filtration rate [eGFR] >30 mL/min/1.73 m2) while in ILLUMINATE-C, individuals with moderately or severely reduced GFR including individuals with kidney failure on hemodialysis were enrolled. In ILLUMINATE-A, 39 individuals were randomized 2:1 to lumasiran or placebo for 6 months. The primary endpoint was the percent change in 24-hour urinary oxalate excretion from baseline to month 6. The percent reduction in 24-hour urinary oxalate from baseline to month 6 was -65% and -12% in the lumasiran and placebo groups, respectively, with a between-group mean difference of 53% (95% confidence interval [CI], 45% to 62%; p<.0001). A similar effect was seen in individuals with high baseline urinary oxalate values, and approximately half of individuals who received lumasiran achieved normal urinary oxalate values by month 6. In ILLUMINATE-B, 18 individuals were treated with lumasiran. The primary endpoint was the percent change in spot urinary oxalate-to-creatinine ratio from baseline to month 6. Lumasiran demonstrated a percent reduction in spot urinary oxalate-to creatinine ratio from baseline of -71% (95% CI, -77% to -65%) at 6 months. Results at 1 year follow-up showed that treatment effects were sustained. The magnitude of the reduction and the time course were consistent with findings in ILLUMINATE-A. In ILLUMINATE-C, 21 individuals were treated with lumasiran. The primary endpoint was the percent change in plasma oxalate levels from baseline to month 6. Lumasiran demonstrated a percent reduction in plasma oxalate levels of -33% (95% CI, -82% to 15%) and -42% (95% CI, -51% to -34%) in individuals who did not require dialysis at the time of study enrollment and individuals who were on a stable regimen of hemodialysis. The major limitation is the lack of data on health outcomes such as renal stones, nephrocalcinosis, and renal failure since neither trial was powered to assess these health outcomes. However, use of urinary oxalate as a surrogate for health outcomes in the pivotal trials may be justified based on the knowledge of the pathophysiology of the disease and the causal role of urinary oxalate in kidney stone formation, nephrocalcinosis, and loss of kidney function. Further, the consistency and size of treatment effect across 3 trials are indicative of the potential for a clinical benefit over the long term. Lumasiran was generally well-tolerated in all 3 studies.


References

Alnylam Pharmaceuticals, Inc. Oxlumo (lumasiran) injection, for subcutaneous use. Cambridge, MA: Alnylam; revised October 2022.


Alnylam Pharmaceuticals. Oxlumo (lumasiran). Product Fact Sheet. Cambridge, MA: Alnylam Pharmaceuticals; 2020. Available at: https://www.alnylam.com/wp-content/uploads/pdfs/OXLUMO-Product-Fact-Sheet.pdf. Accessed December 8, 2021.


American Academy of Pediatrics Steering Committee on Quality Improvement and Management. Classifying recommendations for clinical practice guidelines. Pediatrics. Sep 2004; 114(3): 874-7. PMID 15342869.


Bergstralh EJ, Monico CG, Lieske JC, et al. Transplantation outcomes in primary hyperoxaluria. Am J Transplant. Nov 2010; 10(11): 2493-501. PMID 20849551.


Bhasin B, Ürekli HM, Atta MG. Primary and secondary hyperoxaluria: Understanding the enigma. World J Nephrol. May 06 2015; 4(2): 235-44. PMID 25949937.


Center for Drug Evaluation and Research. Application Number: 214103Orig1s000 Integrated Review for NDA 214103 Oxlumo (lumasiran). Available at https://www.accessdata.fda.gov/drugsatfda_docs/nda/2020/214103Orig1s000IntegratedR.pdf. Accessed on April 25, 2022.


Chlebeck PT, Milliner DS, Smith LH. Long-term prognosis in primary hyperoxaluria type II (L-glyceric aciduria). Am J Kidney Dis. Feb 1994; 23(2): 255-9. PMID 8311084.


Cochat P, Deloraine A, Rotily M, et al. Epidemiology of primary hyperoxaluria type 1. Société de Néphrologie and the Société de Néphrologie Pédiatrique. Nephrol Dial Transplant. 1995; 10 Suppl 8: 3-7. PMID 8592622.


Cochat P, Hulton SA, Acquaviva C, et al. Primary hyperoxaluria Type 1: indications for screening and guidance for diagnosis and treatment. Nephrol Dial Transplant. May 2012; 27(5): 1729-36. PMID 22547750.


Cochat P, Koch Nogueira PC, Mahmoud MA, et al. Primary hyperoxaluria in infants: medical, ethical, and economic issues. J Pediatr. Dec 1999; 135(6): 746-50. PMID 10586179.


Cochat P, Rumsby G. Primary hyperoxaluria. N Engl J Med. Aug 15 2013; 369(7): 649-58. PMID 23944302.


Dick PT, Shuckett BM, Tang B, et al. Observer reliability in grading nephrocalcinosis on ultrasound examinations in children. Pediatr Radiol. Jan 1999; 29(1): 68-72. PMID 9880623.


Frishberg Y, Deschênes G, Groothoff JW, et al. Phase 1/2 Study of Lumasiran for Treatment of Primary Hyperoxaluria Type 1: A Placebo-Controlled Randomized Clinical Trial. Clin J Am Soc Nephrol. Jul 2021; 16(7): 1025-1036. PMID 33985991.


Galanti M, Contreras A. Excellent renal function and reversal of nephrocalcinosis 8 years after isolated liver transplantation in an infant with primary hyperoxaluria type 1. Pediatr Nephrol. Nov 2010; 25(11): 2359-62. PMID 20628764.


Garrelfs SF, Frishberg Y, Hulton SA, et al; ILLUMINATE-A Collaborators. Lumasiran, an RNAi therapeutic for primary hyperoxaluria type 1. N Engl J Med. 2021;384(13):1216-1226.


Garrelfs SF, Rumsby G, Peters-Sengers H, et al. Patients with primary hyperoxaluria type 2 have significant morbidity and require careful follow-up. Kidney Int. Dec 2019; 96(6): 1389-1399. PMID 31685312.


Groothoff JW, Metry E, Deesker L, et al. Clinical practice recommendations for primary hyperoxaluria: an expert consensus statement from ERKNet and OxalEurope. Nat Rev Nephrol. Mar 2023; 19(3): 194-211. PMID 36604599.


Harambat J, Fargue S, Acquaviva C, et al. Genotype-phenotype correlation in primary hyperoxaluria type 1: the p.Gly170Arg AGXT mutation is associated with a better outcome. Kidney Int. Mar 2010; 77(5): 443-9. PMID 20016466.


Harambat J, Fargue S, Bacchetta J, et al. Primary hyperoxaluria. Int J Nephrol. 2011; 2011: 864580. PMID 21748001.


Hayes W, Sas DJ, Magen D, et al. Efficacy and safety of lumasiran for infants and young children with primary hyperoxaluria type 1: 12-month analysis of the phase 3 ILLUMINATE-B trial. Pediatr Nephrol. Apr 2023; 38(4): 1075-1086. PMID 35913563.


Hopp K, Cogal AG, Bergstralh EJ, et al. Phenotype-Genotype Correlations and Estimated Carrier Frequencies of Primary Hyperoxaluria. J Am Soc Nephrol. Oct 2015; 26(10): 2559-70. PMID 25644115.


Hoppe B, Beck BB, Milliner DS. The primary hyperoxalurias. Kidney Int. Jun 2009; 75(12): 1264-1271. PMID 19225556.


Hoppe B, Graf D, Offner G, et al. Oxalate elimination via hemodialysis or peritoneal dialysis in children with chronic renal failure. Pediatr Nephrol. Aug 1996; 10(4): 488-92. PMID 8865249.


Hoppe B, Kemper MJ, Bökenkamp A, et al. Plasma calcium oxalate supersaturation in children with primary hyperoxaluria and end-stage renal failure. Kidney Int. Jul 1999; 56(1): 268-74. PMID 10411702.


Hoppe B, Kemper MJ, Bökenkamp A, et al. Plasma calcium-oxalate saturation in children with renal insufficiency and in children with primary hyperoxaluria. Kidney Int. Sep 1998; 54(3): 921-5. PMID 9734617.


Hoppe B, Langman CB. A United States survey on diagnosis, treatment, and outcome of primary hyperoxaluria. Pediatr Nephrol. Oct 2003; 18(10): 986-91. PMID 12920626.


Hoppe B. An update on primary hyperoxaluria. Nat Rev Nephrol. Jun 12 2012; 8(8): 467-75. PMID 22688746.


Khorsandi SE, Samyn M, Hassan A, et al. An institutional experience of pre-emptive liver transplantation for pediatric primary hyperoxaluria type 1. Pediatr Transplant. Jun 2016; 20(4): 523-9. PMID 27061278.


Kopp N, Leumann E. Changing pattern of primary hyperoxaluria in Switzerland. Nephrol Dial Transplant. Dec 1995; 10(12): 2224-7. PMID 8808215.


Liebow A, Li X, Racie T, et al. An investigational RNAi therapeutic targeting glycolate oxidase reduces oxalate production in models of primary hyperoxaluria. J Am Soc Nephrol. 2017;28(2):494-503.


Marangella M, Petrarulo M, Cosseddu D, et al. Oxalate balance studies in patients on hemodialysis for type I primary hyperoxaluria. Am J Kidney Dis. Jun 1992; 19(6): 546-53. PMID 1595703.


Marangella M, Petrarulo M, Vitale C, et al. Serum calcium oxalate saturation in patients on maintenance haemodialysis for primary hyperoxaluria or oxalosis-unrelated renal diseases. Clin Sci (Lond). Oct 1991; 81(4): 483-90. PMID 1657494.


McGregor TL, Hunt KA, Yee E, et al. Characterising a healthy adult with a rare HAO1 knockout to support a therapeutic strategy for primary hyperoxaluria. Elife. 2020;9:e54363.


Michael M, Groothoff JW, Shasha-Lavsky H, et al. Lumasiran for Advanced Primary Hyperoxaluria Type 1: Phase 3 ILLUMINATE-C Trial. Am J Kidney Dis. Feb 2023; 81(2): 145-155.e1. PMID 35843439.


Milliner DS, Harris PC, Cogal AG, et al. Primary Hyperoxaluria Type 1. 2002 Jun 19 [Updated 2017 Nov 30]. In: Adam MP, Ardinger HH, Pagon RA, et al., editors. GeneReviews [Internet]. Seattle (WA): University of Washington, Seattle; 1993-2021.


Available from: https://www.ncbi.nlm.nih.gov/books/NBK1283/. Accessed April 25, 2022.


Milliner DS. The primary hyperoxalurias: An algorithm for diagnosis. Am J Nephrol. 2005;25(2):154-160.


Niaudet P. Primary hyperoxaluria. UpToDate [online serial]. Waltham, MA: UpToDate; reviewed June 2022.


Nolkemper D, Kemper MJ, Burdelski M, et al. Long-term results of pre-emptive liver transplantation in primary hyperoxaluria type 1. Pediatr Transplant. Aug 2000; 4(3): 177-81. PMID 10933316.


Prescribing Label: OXLUMO (lumasiran) injection, for subcutaneous use. Available at https://www.alnylam.com/wp-content/uploads/pdfs/OXLUMO-Prescribing-Information.pdf. Accessed on April 25, 2022.


Rumsby G, Williams E, Coulter-Mackie M. Evaluation of mutation screening as a first line test for the diagnosis of the primary hyperoxalurias. Kidney Int. Sep 2004; 66(3): 959-63. PMID 15327387.


Rumsby, G. Primary hyperoxaluria Type 2. Gene Reviews. http://www.ncbi.nlm.nih.gov/bookshelf/br.fcgi?book=gene&part=ph2. Accessed April 25, 2022.


Sas DJ, Magen D, Hayes W, et al. Phase 3 trial of lumasiran for primary hyperoxaluria type 1: A new RNAi therapeutic in infants and young children. Genet Med. Mar 2022; 24(3): 654-662. PMID 34906487.


Sikora P, von Unruh GE, Beck B, et al. [13C2]oxalate absorption in children with idiopathic calcium oxalate urolithiasis or primary hyperoxaluria. Kidney Int. May 2008; 73(10): 1181-6. PMID 18337715.


U.S. Food and Drug Administration (FDA). FDA approves first drug to treat rare metabolic disorder. FDA News Release. Silver Spring, MD: FDA; November 23, 2020.


van Woerden CS, Groothoff JW, Wanders RJ, et al. Primary hyperoxaluria type 1 in The Netherlands: prevalence and outcome. Nephrol Dial Transplant. Feb 2003; 18(2): 273-9. PMID 12543880.


Zhao F, Bergstralh EJ, Mehta RA, et al. Predictors of Incident ESRD among Patients with Primary Hyperoxaluria Presenting Prior to Kidney Failure. Clin J Am Soc Nephrol. Jan 07 2016; 11(1): 119-26. PMID 26656319​​.


Coding

CPT Procedure Code Number(s)
N/A

ICD - 10 Procedure Code Number(s)
N/A

ICD - 10 Diagnosis Code Number(s)
E72.53 Primary hyperoxaluria

HCPCS Level II Code Number(s)
​J0224 Injection, lumasiran, 0.5 mg

Revenue Code Number(s)
N/A


Coding and Billing Requirements


Policy History

Revisions From 08.01.74:
01/02/2024
The following new policy has been developed to communicate the Company’s coverage criteria for Lumasiran (Oxlumo™)​. The policy will become effective on 01/02/2024.

1/2/2024
1/2/2024
08.01.74
Medical Policy Bulletin
Commercial
No