Randomized placebo-controlled dose-ranging and pharmacodynamics study of roxadustat (FG-4592) to treat anemia in nondialysis-dependent chronic kidney disease (NDD-CKD) patients

Anatole Besarab, Robert Provenzano, Joachim Hertel, Raja Zabaneh, Stephen J Klaus, Tyson Lee, Robert Leong, Stefan Hemmerich, Kin-Hung Peony Yu, Thomas B Neff, Anatole Besarab, Robert Provenzano, Joachim Hertel, Raja Zabaneh, Stephen J Klaus, Tyson Lee, Robert Leong, Stefan Hemmerich, Kin-Hung Peony Yu, Thomas B Neff

Abstract

Background: Roxadustat (FG-4592) is an oral hypoxia-inducible factor prolyl hydroxylase inhibitor that stimulates erythropoiesis. This Phase 2a study tested efficacy (Hb response) and safety of roxadustat in anemic nondialysis-dependent chronic kidney disease (NDD-CKD) subjects.

Methods: NDD-CKD subjects with hemoglobin (Hb) ≤11.0 g/dL were sequentially enrolled into four dose cohorts and randomized to roxadustat or placebo two times weekly (BIW) or three times weekly (TIW) for 4 weeks, in an approximate roxadustat:placebo ratio of 3:1. Efficacy was assessed by (i) mean Hb change (ΔHb) from baseline (BL) and (ii) proportion of Hb responders (ΔHb ≥ 1.0 g/dL). Pharmacodynamic evaluation was performed in a subset of subjects. Safety was evaluated by adverse event frequency/severity.

Results: Of 116 subjects receiving treatment, 104 completed 4 weeks of dosing and 96 were evaluable for efficacy. BL characteristics for roxadustat and placebo groups were comparable. In roxadustat-treated subjects, Hb levels increased from BL in a dose-related manner in the 0.7, 1.0, 1.5 and 2.0 mg/kg groups. Maximum ΔHb within the first 6 weeks was significantly higher in the 1.5 and 2.0 mg/kg groups than in the placebo subjects. Hb responder rates were dose dependent and ranged from 30% in the 0.7 mg/kg BIW group to 100% in the 2.0 mg/kg BIW and TIW groups versus 13% in placebo.

Conclusions: Roxadustat transiently and moderately increased endogenous erythropoietin and reduced hepcidin. Adverse events were similar in the roxadustat and placebo groups. Roxadustat produced dose-dependent increases in blood Hb among anemic NDD-CKD patients in a placebo-controlled trial.

Clinical trials registration: Clintrials.gov #NCT00761657.

Keywords: HIF-PHI; anemia; chronic kidney disease; erythropoietin; hepcidin.

© The Author 2015. Published by Oxford University Press on behalf of ERA-EDTA.

Figures

FIGURE 1:
FIGURE 1:
Patient disposition. *The AEs in the roxadustat arm were acute prostatitis (in the 1.0 mg/kg TIW group) and elevated liver enzymes (in the 2.0 mg/kg BIW group). One placebo patient was discontinued because of SAEs of acute pericarditis and renal failure.
FIGURE 2:
FIGURE 2:
Mean maximum change from BL in Hb (ΔHbmax) and % subjects achieved Hb response, defined as Hb increase by ≥1 g/dL (EE population). Mean (SD) BL Hb was 10.1 (0.7) g/dL for roxadustat subjects and 10.1 (0.6) g/dL for placebo subjects. Pooled placebo data used. Time to response was estimated using the Kaplan–Meier method, estimable for groups with >50% response. Nonresponders were censored at Day 42. *From an intergroup t-test compared with placebo; n.s.: not significant.
FIGURE 3:
FIGURE 3:
Mean change from BL in Hb (ΔHb) in TIW cohorts (EE population). Mean (SD) BL Hb was 10.1 (0.7) g/dL for roxadustat TIW subjects and 10.1 (0.6) g/dL for placebo subjects. Last-observation-carried-forward (LOCF) method was used to impute missing values. *P t-tests comparing roxadustat change from BL with placebo change from BL. End of treatment (EOT) for TIW was Day 26.
FIGURE 4:
FIGURE 4:
Changes in median plasma EPO on first and final days of treatment. Data are for the PK/PD population (subjects with complete dataset only).
FIGURE 5:
FIGURE 5:
Mean change from BL in serum hepcidin (EE population). Data from BIW and TIW groups were pooled for each dose level. Serum hepcidin was not measured in the roxadustat 1.0 mg/kg dose group. LOCF method was used to impute missing data. *P = 0.048, **P = 0.0013, intergroup two-sample t-tests comparing roxadustat change from BL with placebo change from BL. EOT was Day 29 (BIW) or Day 26 (TIW).

References

    1. Collins AJ, Ma JZ, Xia A et al. . Trends in anemia treatment with erythropoietin usage and patient outcomes. Am J Kidney Dis 1998; 32: S133–S141
    1. Coresh J, Selvin E, Stevens LA et al. . Prevalence of chronic kidney disease in the United States. JAMA 2007; 298: 2038–2047
    1. Eschbach JW, Egrie JC, Downing MR et al. . Correction of the anemia of end-stage renal disease with recombinant human erythropoietin. Results of a combined phase I and II clinical trial. N Engl J Med 1987; 316: 73–78
    1. Regidor DL, Kopple JD, Kovesdy CP et al. . Associations between changes in hemoglobin and administered erythropoiesis-stimulating agent and survival in hemodialysis patients. J Am Soc Nephrol 2006; 17: 1181–1191
    1. Revicki DA, Brown RE, Feeny DH et al. . Health-related quality of life associated with recombinant human erythropoietin therapy for predialysis chronic renal disease patients. Am J Kidney Dis 1995; 25: 548–554
    1. Benz R, Schmidt R, Kelly K et al. . Epoetin alfa once every 2 weeks is effective for initiation of treatment of anemia of chronic kidney disease. Clin J Am Soc Nephrol 2007; 2: 215–221
    1. U.S. Renal Data System. 2013 Annual Data Report: Atlas of End-Stage Renal Disease in the United States. Bethesda, MD: National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases; (1 October 2014, date last accessed)
    1. Minutolo R, Locatelli F, Gallieni M et al. . Anaemia management in non-dialysis chronic kidney disease (CKD) patients: a multicentre prospective study in renal clinics. Nephrol Dial Transplant 2013; 28: 3035–3045
    1. Yan G, Cheung AK, Ma JZ et al. . The associations between race and geographic area and quality-of-care indicators in patients approaching ESRD. Clin J Am Soc Nephrol 2013; 8: 610–618
    1. Valderrabano F, Horl WH, Macdougall IC et al. . PRE-dialysis survey on anaemia management. Nephrol Dial Transplant 2003; 18: 89–100
    1. Perez-Garcia R, Martin-Malo A, Fort J et al. . Baseline characteristics of an incident haemodialysis population in Spain: results from ANSWER—a multicentre, prospective, observational cohort study. Nephrol Dial Transplant 2009; 24: 578–588
    1. Pfeffer MA, Burdmann EA, Chen CY et al. . A trial of darbepoetin alfa in type 2 diabetes and chronic kidney disease. N Engl J Med 2009; 361: 2019–2032
    1. Singh AK, Szczech L, Tang KL et al. . Correction of anemia with epoetin alfa in chronic kidney disease. N Engl J Med 2006; 355: 2085–2098
    1. Epogen® Package Insert, 2009. (20 March 2015, date last accessed)
    1. Gill KS, Muntner P, Lafayette RA et al. . Red blood cell transfusion use in patients with chronic kidney disease. Nephrol Dial Transplant 2013; 28: 1504–1515
    1. Seliger S, Fox KM, Gandra SR et al. . Timing of erythropoiesis-stimulating agent initiation and adverse outcomes in nondialysis CKD: a propensity-matched observational study. Clin J Am Soc Nephrol 2010; 5: 882–888
    1. Levin A. The treatment of anemia in chronic kidney disease: understandings in 2006. Curr Opin Nephrol Hypertens 2007; 16: 267–271
    1. Mohanram A, Zhang Z, Shahinfar S et al. . Anemia and end-stage renal disease in patients with type 2 diabetes and nephropathy. Kidney Int 2004; 66: 1131–1138
    1. Karpinski M, Pochinco D, Dembinski I et al. . Leukocyte reduction of red blood cell transfusions does not decrease allosensitization rates in potential kidney transplant candidates. J Am Soc Nephrol 2004; 15: 818–824
    1. Obrador GT, Macdougall IC. Effect of red cell transfusions on future kidney transplantation. Clin J Am Soc Nephrol 2013; 8: 852–860
    1. Macdougall IC. Recent advances in erythropoietic agents in renal anemia. Semin Nephrol 2006; 26: 313–318
    1. Haase VH. Hypoxic regulation of erythropoiesis and iron metabolism. Am J Physiol Renal Physiol 2010; 299: F1–F13
    1. Levey AS, Bosch JP, Lewis JB et al. . A more accurate method to estimate glomerular filtration rate from serum creatinine: a new prediction equation. Modification of Diet in Renal Disease Study Group. Ann Intern Med 1999; 130: 461–470
    1. Macdougall IC, Walker R, Provenzano R et al. . C.E.R.A. corrects anemia in patients with chronic kidney disease not on dialysis: results of a randomized clinical trial. Clin J Am Soc Nephrol 2008; 3: 337–347
    1. Ross SP, McCrea JB, Besarab A. Erythropoietin response to blood loss in hemodialysis patients is blunted but preserved. ASAIO J 1994; 40: M880–M885
    1. Kato A, Hishida A, Kumagai H et al. . Erythropoietin production in patients with chronic renal failure. Ren Fail 1994; 16: 645–651
    1. Milledge JS, Cotes PM. Serum erythropoietin in humans at high altitude and its relation to plasma renin. J Appl Physiol 1985; 59: 360–364
    1. Cheung WK, Goon BL, Guilfoyle MC et al. . Pharmacokinetics and pharmacodynamics of recombinant human erythropoietin after single and multiple subcutaneous doses to healthy subjects. Clin Pharmacol Ther 1998; 64: 412–423
    1. Fishbane S, Besarab A. Mechanism of increased mortality risk with erythropoietin treatment to higher hemoglobin targets. Clin J Am Soc Nephrol 2007; 2: 1274–1282
    1. Szczech LA, Barnhart HX, Inrig JK et al. . Secondary analysis of the CHOIR trial epoetin-alpha dose and achieved hemoglobin outcomes. Kidney Int 2008; 74: 791–798
    1. Zhang Y, Thamer M, Stefanik K et al. . Epoetin requirements predict mortality in hemodialysis patients. Am J Kidney Dis 2004; 44: 866–876
    1. Erslev AJ. Erythropoietin. N Engl J Med 1991; 324: 1339–1344
    1. Arcasoy MO. The non-haematopoietic biological effects of erythropoietin. Br J Haematol 2008; 141: 14–31
    1. Spinowitz BS, Kausz AT, Baptista J et al. . Ferumoxytol for treating iron deficiency anemia in CKD. J Am Soc Nephrol 2008; 19: 1599–1605
    1. Locatelli F, Olivares J, Walker R et al. . Novel erythropoiesis stimulating protein for treatment of anemia in chronic renal insufficiency. Kidney Int 2001; 60: 741–747
    1. Macdougall IC, Wiecek A, Tucker B et al. . Dose-finding study of peginesatide for anemia correction in chronic kidney disease patients. Clin J Am Soc Nephrol 2011; 6: 2579–2586
    1. Roger SD, Locatelli F, Woitas RP et al. . C.E.R.A. once every 4 weeks corrects anaemia and maintains haemoglobin in patients with chronic kidney disease not on dialysis. Nephrol Dial Transplant 2011; 26: 3980–3986
    1. Nemeth E, Ganz T. Regulation of iron metabolism by hepcidin. Annu Rev Nutr 2006; 26: 323–342
    1. Peyssonnaux C, Zinkernagel AS, Schuepbach RA et al. . Regulation of iron homeostasis by the hypoxia-inducible transcription factors (HIFs). J Clin Invest 2007; 117: 1926–1932
    1. Shah YM, Matsubara T, Ito S et al. . Intestinal hypoxia-inducible transcription factors are essential for iron absorption following iron deficiency. Cell Metab 2009; 9: 152–164
    1. Anderson ER, Xue X, Shah YM. Intestinal hypoxia-inducible factor-2α (HIF-2α) is critical for efficient erythropoiesis. J Biol Chem 2011; 286: 19533–19540

Source: PubMed

3
Abonnere