Among patients with peritoneal dialysis dependent

Similar documents
Intravenous Iron Requirement in Adult Hemodialysis Patients

The safety and efficacy of an accelerated iron sucrose dosing regimen in patients with chronic kidney disease

Title: Parenteral Iron Therapy for Anemia: A Clinical and Cost-Effectiveness Review

ADVANCES. Annual reports from the Centers for. In Anemia Management. Anemia Management in the United States: Is There Opportunity for Improvement?

ferric carboxymaltose 50mg iron/ml solution for injection/infusion (Ferinject ) SMC No. (463/08) Vifor Pharmaceuticals

The safety and efficacy of ferumoxytol therapy in anemic chronic kidney disease patients

ferric carboxymaltose 50mg iron/ml solution for injection/infusion (Ferinject ) SMC No. (463/08) Vifor Pharma UK Ltd

Update on Chemotherapy- Induced Anemia and Neutropenia Therapies

K atching Up with KDOQI: Clinical Practice Guidelines & Clinical Practice Recommendations for Anemia of Chronic Kidney Disease 2006

Epogen / Procrit. Epogen / Procrit (epoetin alfa) Description

The FIND-CKD Study Background Study design (Results)

Iron metabolism anemia and beyond. Jacek Lange Perm, 8 October 2016

The clinical trial information provided in this public disclosure synopsis is supplied for informational purposes only.

Intravenous Iron: A Good Thing Made Better? Marilyn Telen, MD Wellcome Professor of Medicine Duke University

New Aspects to Optimize Epoetin Treatment with Intravenous Iron Therapy in Hemodialysis Patients

Aranesp. Aranesp (darbepoetin alfa) Description

Anemia Management in Peritoneal Dialysis Patients Pranay Kathuria, FACP, FASN

Efficacy and tolerability of oral Sucrosomial Iron in CKD patients with anemia. Ioannis Griveas, MD, PhD

Safety and tolerability of intravenous ferric carboxymaltose in patients with iron deficiency anemia

Guideline Summary NGC-6019

ANEMIA & HEMODIALYSIS

Evaluation of the effect of oral versus intravenous iron treatments on anemia in patients with chronic kidney diseases.

SYNOPSIS. Issue Date: 04 February 2009 Document No.: EDMS -USRA

Published Online 2013 July 24. Research Article

Summary of Recommendation Statements Kidney International Supplements (2012) 2, ; doi: /kisup

The clinical trial information provided in this public disclosure synopsis is supplied for informational purposes only.

Effective Health Care Program

The clinical trial information provided in this public disclosure synopsis is supplied for informational purposes only.

Future Direction of Anemia Management in ESRD. Jay B. Wish, MD 2008 Nephrology Update March 20, 2008

Improvement in Pittsburgh Symptom Score Index After Initiation of Peritoneal Dialysis

IN-CENTER HEMODIALYSIS (HD) CLINICAL PERFORMANCE MEASURES DATA COLLECTION FORM 2006

LVHN Scholarly Works. Lehigh Valley Health Network. Nelson Kopyt DO, FASN, FACP Lehigh Valley Health Network,

Iron deficiency anemia in chronic kidney disease: Uncertainties and cautions

Current situation and future of renal anemia treatment. FRANCESCO LOCATELLI

The Effect of Residual Renal Function at the Initiation of Dialysis on Patient Survival

PERITONEAL DIALYSIS CLINICAL PERFORMANCE MEASURES DATA COLLECTION FORM 2006

Anemia in ESRD patients is effectively treated by the

Regional variability in anaemia management and haemoglobin in the US

EPO e Ferro in Emodialisi: Il PBM al suo esordio. Lucia Del Vecchio. Divisione di Nefrologia e Dialisi Ospedale A. Manzoni, ASST Lecco

Venofer (iron sucrose injection, USP)

Intravenous Iron Does Not Affect the Rate of Decline of Residual Renal Function in Patients on Peritoneal Dialysis

mean hemoglobin 11 g/dl (110 g/l) compared to patients with lower mean hemoglobin values (Table 20).

Once-weekly darbepoetin alfa is as effective as three-times weekly epoetin

PFIZER INC. THERAPEUTIC AREA AND FDA APPROVED INDICATIONS: See USPI

Comment on European Renal Best Practice Position Statement on Anaemia Management in Chronic Kidney Disease.

Clinical Policy: Iron Sucrose (Venofer) Reference Number: CP.PHAR.167

Chapter Five Clinical indicators & preventive health

TITLE: Ferumoxytol versus Other Intravenous Iron Therapies for Anemia: A Review of the Clinical and Cost-effectiveness and Guidelines An Update

Stages of chronic kidney disease

In-House. Solution for Injection A dark brown coloured, slightly viscous solution filled in ampoule.

No Disclosures 03/20/2019. Learning Objectives. Renal Anemia: The Basics

Centocor Ortho Biotech Services, LLC

GUIDELINES FOR ADMINISTRATION OF INTRAVENOUS IRON IN ADULTS WITH CHRONIC KIDNEY DISEASE

Clinical Policy: Ferumoxytol (Feraheme) Reference Number: CP.PHAR.165

Iron Supplementation and Erythropoiesis-Stimulatory Agents in the Treatment of Cancer Anemia

Reference ID:

The efficacy of intravenous darbepoetin alfa administered once every 2 weeks in chronic kidney disease patients on haemodialysis

FDA Advisory Committee Briefing Document

The mortality rate of treated patients with ESRD was 23

See Important Reminder at the end of this policy for important regulatory and legal information.

The Changing Clinical Landscape of Anemia Management in Patients With CKD: An Update From San Diego Presentation 1

Hemodialysis patients with endstage

Anemia Management: Using Epo and Iron

Evidence-based practice in nephrology : Meta-analysis

William E. Strauss *, Naomi V. Dahl, Zhu Li, Gloria Lau and Lee F. Allen

Drugs Used in Anemia

Dear Dr or Infusion Center : Patient name: SS #: Patient address: Patient phone: Date of birth: / /

1 INDICATIONS AND USAGE. 1.1 Limitation of Use FULL PRESCRIBING INFORMATION

Medication Prior Authorization Form

Clinical Policy: Ferric Carboxymaltose (Injectafer) Reference Number: CP.PHAR.234

Iron losses in dialysis patients exceed dietary absorption of

Clinical Performance Goals

Supplementary Online Content

International Journal of Current Research in Chemistry and Pharmaceutical Sciences Volume 1 Issue: Pages: 20-28

Advances in Peritoneal Dialysis, Vol. 23, 2007

Feraheme (ferumoxytol) Injection Coding Tool

OPTA-therapy with iron and erythropoiesis-stimulating agents in chronic kidney disease

CPR 3.5. EVALUATING AND CORRECTING PERSISTENT FAILURE TO REACH OR MAINTAIN INTENDED HB

Conversion Dosing Guide:

Preoperative anemia Common, consequential and correctable in non-emergent surgery By Kathrine Frey, MD

AETNA BETTER HEALTH Prior Authorization guideline for Erythropoiesis Stimulating Agents (ESA)

Study of Management of anemia in Chronic Kidney Disease Patients

NURSE OR PHARMACIST-LED ANEMIA MANAGEMENT PROTOCOL EDUCATIONAL PACKAGE TABLE OF CONTENTS:

Referral Form. Referring physician s phone: Referring physician s fax:

Intravenous iron therapy is required by almost all hemodialysis

Evidence Table. Study Type: Randomized controlled trial. Study Aim: To compare frequent nocturnal hemodialysis and conventional in-center dialysis.

Title:Trends in Anemia Management in US Hemodialysis Patients

NATIONAL QUALITY FORUM Renal EM Submitted Measures

NURSE OR PHARMACIST-LED ANEMIA MANAGEMENT PROTOCOL

Iron Status in Chronic Renal Failure with Anemia

Clinical Performance Goals

The Diabetes Kidney Disease Connection Missouri Foundation for Health February 26, 2009

Transfusion Burden among Patients with Chronic Kidney Disease and Anemia

Changes in anemia management and hemoglobin levels following revision of a bundling policy to incorporate recombinant human erythropoietin

3/21/2017. Solute Clearance and Adequacy Targets in Peritoneal Dialysis. Peritoneal Membrane. Peritoneal Membrane

Long-term iron accumulation in dialysis patients treated with ferric citrate hydrate: a single-center, 80-week retrospective study in Japan

Is Iron Sucrose (Venofer) a Safe Treatment for People with Chronic Kidney Disease?

Effect of oral liposomal iron versus intravenous iron for treatment of iron deficiency anaemia in CKD patients: a randomized trial

Predictors of the response to treatment in anemic hemodialysis patients with high serum ferritin and low transferrin saturation

Transcription:

CJASN epress. Published on March 29, 2006 as doi: 10.2215/CJN.01541005 Effect of Intravenous Iron Sucrose in Peritoneal Dialysis Patients Who Receive Erythropoiesis-Stimulating Agents for Anemia: A Randomized, Controlled Trial Harmeet Singh,* John Reed, Sylvia Noble, Jose L. Cangiano, and David B. Van Wyck; for the United States Iron Sucrose (Venofer) Clinical Trials Group *Western Nephrology & Metabolic Bone Disease, PC, Lakewood, Colorado; Nephrology Associates, Columbus, Mississippi; Northwestern Louisiana Nephrology, Shreveport, Louisiana; Puerto Rico School of Medicine, San Juan, Puerto Rico; and University of Arizona College of Medicine, Tucson, Arizona Although iron therapy is essential to optimize use of erythropoiesis-stimulating agents (ESA), randomized, controlled trials have heretofore been unavailable to evaluate reliably the efficacy of intravenous iron as an adjuvant to ESA treatment in peritoneal dialysis (PD) patients. In a multicenter trial, patients who had anemia, PD-dependent chronic kidney disease, stable ESA therapy, and a broad range of iron status (ferritin <500 ng/ml, transferrin saturation <25%) were randomly assigned to receive either 1gofiron sucrose intravenously in three divided doses (300 mg over 1.5 h on days 1 and 15, 400 mg over 2.5 h on day 29) or no supplemental iron. No serious adverse drug events occurred after intravenous iron administration. The primary end point, peak hemoglobin increase, was higher (1.3 1.1 versus 0.7 1.1, mean SD; P 0.0028), and anemia intervention (transfusion, increase in ESA dose, or intravenous iron therapy not called for in protocol) occurred later (P 0.0137) and less often in intravenous iron treated patients compared with untreated control subjects (one of 66 [1.3%] versus five of 30 [16.7%]). Among patients who did not require intervention, iron-treated patients showed a calculated net ESA dose decrease compared with untreated control subjects. Baseline iron status did not predict responsiveness to intravenous iron therapy. Intravenous iron sucrose is an effective adjunct to ESA therapy in anemic patients with PD-dependent chronic kidney disease and is administered safely as 300 mg over 1.5 h or 400 mg over 2.5 h. Evidence of iron deficiency at baseline is not required to demonstrate intravenous iron efficacy. Clin J Am Soc Nephrol : -,. doi: 10.2215/CJN.01541005 Among patients with peritoneal dialysis dependent chronic kidney disease (PD-CKD), anemia is nearly universal, and use of erythropoiesis-stimulating agents (ESA) is the standard of care. Anemia management in PD-CKD, however, is commonly complicated by iron deficiency. Failure of oral iron supplementation to correct iron deficiency anemia or prevent negative iron balance in patients with PD-CKD is well described (1,2). The alternative, intravenous iron administration, has shown evidence of efficacy in a series of small studies (1 6). The lack of randomized controlled trials (RCT) in PD-CKD, however, leaves the usefulness of intravenous iron as an adjuvant to ESA in these patients untested and uncertain. Confirmation of adjuvant efficacy requires demonstration that, in ESA-treated patients, the proposed adjuvant serves to increase hemoglobin (Hb) without an increase in ESA dose, to decrease ESA doses without changing Hb, or to prevent red cell transfusions or ESA dose increase. For testing the hypothesis Received October 31, 2005. Accepted January 29, 2006. Published online ahead of print. Publication date available at www.cjasn.org. Address correspondence to: Dr. David B. Van Wyck, Kidney Health Institute LLC, 6720 N. Nanini Drive, Tucson, AZ 85704-6128. Phone: 520-906-8262; Fax: 520-498-5027; E-mail: dvanwyck@sprynet.com that an adjuvant is effective in ESA-treated patients, a clinical trial should include three design elements: The ESA dose should be stable before and after randomization to reduce trial outcomes to a single primary end point (change in Hb); patients should be randomly assigned to either adjuvant treatment or no adjuvant treatment to isolate adjuvant effect, if any; and trial duration should be long enough, at least 12 wk, to permit observation of the complete Hb response. To our knowledge, no previous trial has been designed specifically to examine adjuvant efficacy of intravenous iron in ESA-treated patients with PD-CKD. Iron status tests often are used to trigger intravenous iron therapy. In a small, single-arm interventional trial, the sensitivity of transferrin saturation (TSAT) 20% or ferritin 100 ng/ml to predict responsiveness to intravenous iron in anemic patients with PD-CKD was reported as low (7). Detailed information in larger numbers of patients with PD-CKD is needed to elucidate the relationship between baseline levels of iron status tests and likelihood of response to an intravenous iron challenge. Accordingly, we conducted a 12-wk RCT in anemic patients with PD-CKD and compared treatment of anemia with ESA together with intravenous iron sucrose to treatment with ESA alone. We assessed the adjuvant efficacy of intravenous iron, Copyright by the American Society of Nephrology ISSN: 1555-9041/ -0001

2 Clinical Journal of the American Society of Nephrology Clin J Am Soc Nephrol : -, determined the safety of iron sucrose given at 300- and 400-mg doses, and evaluated the relationship between baseline iron indices and likelihood of response to intravenous iron challenge. Materials and Methods Study Design This was an open-label, phase 3, randomized, multicenter trial that was conducted in adherence to the Declaration of Helsinki. Anemic patients who had PD-CKD and required iron supplementation, met all inclusion and exclusion criteria, and had given informed consent were enrolled into the study at 21 study sites. We randomly assigned patients into study groups when they met criteria for randomization, including ESA therapy (darbepoetin or epoetin- ) without dose change for 8 wk, and no parenteral or oral iron for at least 4 wk. Enrolled patients who were not immediately eligible for randomization underwent monthly laboratory examination for up to 6 mo, followed by randomization upon meeting inclusion criteria (Figure 1). Thus, enrollment served to increase the pool of patients who were available for randomization, and randomization could occur as early as1doraslate as 180 d after enrollment. Inclusion Criteria for Enrollment. Criteria for enrollment were distinct from criteria for randomization. Patients were eligible for enrollment when they were older than 18 yr, were able to give informed consent, underwent PD and ESA therapy, and evidenced anemia (Hb 9.5 g/dl and 12.5 g/dl). In Mexican study sites, because of the severity of prevalent anemia, previous ESA use was not required and lower Hb was permitted for enrollment (Hb 8.5 g/dl and 12.5 g/dl); initiation of ESA after enrollment was permitted, but stable ESA dose for 8 wk was required before randomization. Exclusion Criteria for Enrollment. Patients were excluded from enrollment when they had a history of known sensitivity to iron sucrose (patients with intolerances to other forms of iron were permitted to participate); chronic or serious infection, malignancy, or major surgery in the month before enrollment; blood transfusion within the week before enrollment; clinically significant bleeding within 3 mo before enrollment; concomitant severe diseases of the liver or cardiovascular system or severe psychiatric disorders or other conditions that, in the opinion of the investigator, made participation unacceptable; pregnancy or lactation; current treatment for asthma; anticipated surgery that would have required hospitalization during the study period other than vascular access or peritoneal catheter placement; anticipated dialysis or renal transplantation during the study; administration of an investigational drug within 30 d of enrollment; chronic alcoholism or drug abuse within the past 6 mo; and known hemochromatosis or hemosiderosis. Criteria for Randomization. Patients were eligible for randomization when they continued to meet the enrollment criteria and met the following additional criteria: Hb 9.5 and 11.5 g/dl, TSAT 25%, ferritin 500 ng/ml; stable ESA dose for at least 8 wk; and no iron therapy after enrollment or for at least 4 wk before randomization. In Mexican study sites, because of the severity of prevalent anemia, we permitted a lower limit of Hb for randomization (Hb 8.5 and 11.5 g/dl). Criteria for Premature Withdrawal. Premature withdrawal was required when ESRD treatment modality change to renal transplant or hemodialysis (HD) or an intervention for management of anemia was required. We defined an anemia intervention as a red blood cell transfusion, an increase in ESA dose, or iron administration that was not included in the study protocol. We specified that intervention was required when the Hb fell below 9.5 g/dl during enrollment (8.0 g/dl in Mexico) or below 8.5 g/dl after randomization (8.0 g/dl in Mexico). Of course, patients who wished to withdraw from the study could do so at any time without the need to justify the decision, and investigators could withdraw a patient at any time if withdrawal was believed to be in the best interest of the patient. In the analysis of efficacy and safety, we included data for each patient up to the time of withdrawal. Treatment Patients in the intravenous iron treatment arm received iron 1000 mg of iron sucrose intravenously in divided doses over a 28-d period as a 300-mg infusion in 250 ml of 0.9% NaCl given over 1.5 h on days 1 and 15 (approximately 3.3 mg/min) and a 400-mg infusion over 2.5 h (approximately 2.7 mg/min) on day 29. Patients received ESA at the same dose as before randomization, unchanged throughout the study period (days 1 to 71). Patients in the ESA only treatment arm received ESA at the same dose as before randomization, unchanged throughout the study period (days 1 to 71). Randomization Method for Assigning Treatment Group. We first identified patients as new to PD ( 3 mo) or established PD ( 3 mo). We then independently stratified new and established patients by Hb level (11.5 to 10.5 versus 10.4 to 9.5 g/dl; for sites in Mexico, 9.4 to 8.5). Within each resulting combination of strata, we then randomly assigned patients in a 2:1 ratio to study treatment group A (ESA intravenous iron) or B (ESA alone). Figure 1. Study design depicting enrollment and randomization to either intravenous iron treatment or oral iron treatment. Patients who were not eligible immediately upon enrollment were followed for up to 6 mo and randomly assigned when they became eligible. Determination of Efficacy and Safety Efficacy. We defined the primary efficacy end point as the change from baseline to the highest Hb observed at any time between baseline and either the end of study or withdrawal. We determined sample size for this study on the basis of the hypothesis that the difference in mean peak Hb response between treatment groups would be 0.75 g/dl, assuming a common SD no greater than 1.0 g/dl. To have a 95% power to detect that difference required a minimum of 111 randomly assigned patients (74 in group A, 37 in group B). Secondary end points included the observed increase in TSAT and ferritin after treatment and the time to anemia intervention. Laboratory Evaluation. Blood samples for laboratory analyses that were obtained at all appropriate visits were analyzed by a central laboratory. We determined hematologic indices and iron indices at enrollment; at completion of the enrollment period; and on days 15, 29, 43, 57, and 71 after initiation of therapy. We determined the baseline Hb as the final Hb values before randomization. We determined routine

Clin J Am Soc Nephrol : -, Intravenous Iron in PD Patients Who Receive ESA Therapy 3 clinical chemistry at enrollment and at day 71 or study completion. Iron indices included serum iron, total iron binding capacity, TSAT, and ferritin. Safety (Adverse Events). We monitored BP and recorded adverse events in all patients before, during, and for 20 min after administration of intravenous iron. We asked all patients to report any untoward medical event at the onset and queried each patient at each study visit. We recorded adverse events from the day of consent through the completion of the study (day 71) or 30 d after the last dose of study drug, whichever was later. Investigators provided the date of onset, severity, the relationship to study drug, the date of resolution (or that the event was still continuing), the action taken, and the outcome of the adverse experience. We did not consider worsening anemia or iron deficiency to be adverse events, because these developments became study end points when an anemia intervention was required, as defined above. We classified potential hypersensitivity reactions by grade as events that occurred from the start of drug infusion to 20 min after the completion of the infusion, according to accepted guidelines (National Cancer Institute Common Toxicity Criteria Version 2.0; http://ctep. cancer.gov/reporting/ctc.html). Investigators graded events that lacked accepted terminology as mild (did not interfere with patient s usual function), moderate (interfered to some extent), severe (interfered significantly), or life-threatening (resulted in a threat to life or an incapacitating disability). A serious adverse event included any experience that was fatal or life-threatening, resulted in or prolonged inpatient hospitalization, resulted in persistent or significant disability or incapacity, or presented a significant hazard to the patient. Investigators judged relatedness to study drug as none, unlikely, possible, or probable. Statistical Analyses Primary Efficacy End Point. The principal analysis of the primary end point was the unstratified comparison of the peak Hb increase above baseline between the two study arms (group A versus group B) in the intention-to-treat (ITT) population, using a two-step procedure. In the first step, we used a regression model to identify important covariates (including baseline Hb, TSAT, ferritin, reticulocyte count, study center, age, gender, and race) and potential interactions with treatment. In the second step, we included the treatment effect and significant covariates that were identified in the first step in an analysis of covariance model to estimate an overall treatment effect and the P value for between-group testing. Secondary Efficacy End Points. We analyzed secondary end points using the same procedure as that used for primary end points. We calculated median time to peak Hb response and time to intervention using Kaplan-Meier estimates and compared between-group results using Wald 2 P values based on the Cox proportional hazard model with effects for treatment and baseline Hb. Clinical Utility of Iron Status Tests. To evaluate the clinical utility of iron status tests to discriminate between iron-responsive and iron-nonresponsive patients, we calculated the sensitivity and the specificity of iron status tests at multiple cutoff values, computed receiveroperator characteristic (ROC) curves for each variable, and compared area under the ROC curve against 0.5 (corresponding to the line of no discrimination), using statistical software (Analyze-it version 1.71; Analyze-it Software, Ltd, Leeds, UK). We examined results only for patients in group A and considered true positive (iron responsive) to be a patient who demonstrated a Hb 1.0 g/dl after intravenous iron treatment. Results Patient Disposition A total of 188 patients were enrolled. Time to randomization did not differ between groups (47.2 45.0 versus 29.8 32.9 d, group A versus group B, mean SD). A total of 126 patients were randomly assigned at 27 centers (mean 4.7 patients per center; range 1 to 27) to receive ESA plus iron sucrose (group A: 80 patients) or ESA alone (group B: 46 patients). Of these 126 patients, five were randomly assigned to group A but were discontinued from the study before dosing (Figure 2). Of these five patients, two were lost to follow-up, two were withdrawn as a result of acute adverse events, and one had an unstable ESA dose before randomization. All five patients were excluded from the evaluation of both safety and efficacy. Therefore, the safety-evaluable population included a total of 121 patients who received at least one dose of iron: 75 patients in group A and 46 in group B. The ITT population, which formed the basis for efficacy analysis, included all safety-evaluable patients who had efficacy data after baseline, except for nine patients in group A and eight in group B who were subsequently found to have been randomly assigned despite ESA dose changes within 8 wk before randomization. Efficacy Among the 96 patients in the ITT population, there were no significant differences at baseline between patients in group A (n 66) and those in group B (n 30), except for TSAT (Table 1). Patients in group B showed a statistically significantly lower mean TSAT value at baseline (16.8%) than those in group A (19.8%). Anemia Intervention. One of 66 patients (1.3%) in group A and five of 30 patients (16.7%) in group B required anemia intervention, defined by protocol as an increase in ESA dose, administration of nonprotocol intravenous iron, or red blood cell transfusion. Median time to anemia intervention was significantly shorter among patients without iron treatment than with intravenous iron treatment (34 versus 59 d; Wald 2 P 0.0137; Figure 3). Exploratory analysis revealed that ESA dose decreases occurred in selected patients, contrary to study protocol, in response to elevated Hb. Eleven (16.7%) of 66 patients in group A and three (10.0%) of 33 patients in group B experienced Hb 13.0 g/dl. Eight ESA dose decreases occurred in Figure 2. Disposition of study patients after screening. We examined safety in the safety-evaluable population and efficacy in the intent-to-treat (ITT) population.

4 Clinical Journal of the American Society of Nephrology Clin J Am Soc Nephrol : -, Table 1. Baseline demographic and laboratory values among intention-to-treat patients in intravenous iron and oral iron treatment groups a Baseline Results Group A (ESA IV Iron; n 66) Treatment Group Group B (ESA Alone; n 30) P Demographics age (y) 52.3 55.7 0.9825 race (white/black/hispanic/asian) 27/13/22/4 6/4/17/3 0.0520 gender (male/female) 42/24 14/16 0.1261 epoetin- (IU/wk) 11,681 7,932 0.1012 weight (kg) 79.2 75.4 0.8341 Laboratory Hb (g/dl) 10.6 10.5 0.7821 TSAT (%) 19.8 16.8 0.0043 ferritin (ng/ml) 167.5 194.2 0.2975 TSAT 20% and ferritin 100 ng/ml (n) 12 5 1.0000 previous iron intolerance (dextran/gluconate) 2/2 0/0 0.3059 a Hb, hemoglobin; IV, intravenous; TSAT, transferrin saturation. Figure 3. Effect of treatment on percentage of patients who remained without anemia intervention, including erythropoiesis-stimulating agent (ESA) dose increase, added intravenous iron, or transfusion for low hemoglobin (Hb) during the trial (top) or ESA dose decrease for elevated Hb (bottom). Results are shown for ITT patients. P values are based on the Cox proportional hazard model with effects for treatment and baseline Hb. seven (10.6%) of 66 patients in group A, and two dose decreases occurred in two (6.1%) of 33 patients in group B. Median time to ESA dose decrease was no different in group A than in group B patients (33 versus 36 d, respectively; P 0.5087; Figure 3). However, the magnitude of dose decrease was greater among group A than group B patients. We converted darbepoetin doses if any to epoetin- equivalents by multiplying by 200, then for each patient, we subtracted actual doses given during the trial from those that would have been given had ESA dosing remained unchanged. We calculated that the resulting epoetin- dose decrease totaled 260,528 IU in group A and 65,429 IU in group B. Therefore, the net between-group ESA dose difference that was associated with intravenous iron therapy was 195,099 IU, or 2,956 IU per group A patient. We did not calculate added ESA requirements that were incurred as a result of dose increases, because patients with ESA dose increases were withdrawn from the study. Hb Response. Patients in group A showed a higher peak Hb increase, the primary end point, than did those in group B (1.3 versus 0.6; difference 0.7 [95% confidence interval 0.3 to 1.2]; P 0.0028). Hb increase from baseline was significant at each interval after baseline in group A. We observed no significant increase in Hb at any interval in group B (Figure 4). When compared with patients who were assigned to group B, a greater proportion of patients who were assigned to group A showed an increase in Hb 1.0 g/dl (59.1 versus 33.3%; P 0.0273). Among patients who completed the study, median time to peak Hb response did not differ between groups (48.3 versus 51.4 d). Baseline status, including age, race, gender, Hb, TSAT, and ferritin, showed no significant effect on peak Hb increase or proportion of patients with Hb increase 1 g/dl. Study site showed no discernible effect on efficacy results, whether all patients among all sites were examined by the a priori statistical plan or US patients were compared with Mexican patients by exploratory analysis. When we compared Mexican study patients with those in the United States, we found neither an overall country effect on Hb outcomes between groups A and B nor a selective country effect within either group A or group B. When we removed Mexican sites from analysis, US-only patients showed the relative between-group

Clin J Am Soc Nephrol : -, Intravenous Iron in PD Patients Who Receive ESA Therapy 5 Figure 4. Hb response in anemic peritoneal dialysis (PD) patients after intravenous iron compared with no iron therapy. Proportion of patients who achieved Hb 1.0 g/dl (top) and actual Hb (bottom; mean 95% confidence interval [CI] of mean). Between treatment groups, significant differences were present for cumulative response by day 71 ( P 0.05). Within each group, significant differences in Hb from baseline are shown (*P 0.05, **P 0.001, ***P 0.0001). efficacy effects that were seen among all patients combined, but smaller sample sizes (n 47 and 15, groups A and B, respectively) rendered comparisons not statistically significant. Iron Stores and Iron Adequacy for Erythropoiesis, Response to Treatment. Serum ferritin increased significantly from baseline only in group A, and differences between groups were highly significant (Figure 5). Peak ferritin values attained were higher in group A than in group B (P 0.0001) and correlated directly with baseline Hb (P 0.0409) and baseline ferritin (P 0.0004). TSAT increased in both groups; differences between groups narrowly failed to achieve statistical significance. However, peak TSAT values where significantly higher in group A than in group B (P 0.0098) and correlated directly with both baseline Hb (P 0.0007) and baseline ferritin (P 0.0120). Clinical Utility of Baseline Iron Indices. The clinical utility of baseline iron status tests to discriminate between patients who are iron responsive, defined as a Hb after intravenous iron therapy 1g/dl, and those who are iron nonresponsive, defined as a Hb 1 g/dl, was weak. Baseline TSAT and ferritin showed no diagnostic utility at any cutoff value (Figure 6). Safety Extent of Exposure. Among the 75 patients in group A, five had a history of intolerance to an intravenous iron agent (two to iron dextran, two to ferric gluconate, and one to both). Group A patients received 149 doses of iron sucrose as 300 mg over 90 min and 70 doses of iron sucrose as 400 mg over 2.5 h. The mean cumulative per-patient dose of intravenous iron sucrose that was administered in the safety population was 970.9 mg (95% confidence of the mean 944.4 to 997.5 mg). Figure 5. Change in iron status indicators in patients after intravenous iron compared with oral iron therapy (mean 95% CI of mean). Between-group differences were significant for ferritin at all time periods (P 0.0001) and transferrin saturation ( TSAT) at day 14 (P 0.0314). Within each group, significant differences from baseline are shown (*P 0.05, ***P 0.001). Figure 6. Receiver operating characteristic (ROC) curves examining the utility of iron status tests to discriminate between iron responsive and iron-nonresponsive study patients. We defined iron responsive as showing a Hb 1 g/dl after intravenous iron treatment. Results include only patients in group A, the intravenous iron treatment group. No test showed discriminating utility at any cutoff value (TSAT area under the curve [AUC] 0.539 [95% CI 0.299 to 0.779]; ferritin AUC 0.507 [95% CI 0.272 to 0.742]). Adverse Events. There were no serious adverse drug events (ADE). Sixty-two patients in group A and 26 patients in group B completed the study. An additional 22 patients (12 in group A, 10 in group B) discontinued for reasons other than intervention (Table 2). Three patients in group A discontinued as a result of adverse events; of these, one experienced an adverse event that was considered study drug related. This patient experienced swelling of the feet and pruritus 30 to 45

6 Clinical Journal of the American Society of Nephrology Clin J Am Soc Nephrol : -, Table 2. Reason for discontinuation other than anemia intervention, by treatment group a Group A (ESA IV iron) Group B (ESA Alone) Total patients treated (safety assessable) 75 (100%) 46 (100%) Reasons for discontinuation adverse event related to study drug 1 (1.3%) 0 adverse event unrelated to study drug 2 (2.7%) 1 (2.2%) selection criteria/study compliance 3 (4.0%) 3 (6.5%) renal transplant 3 (4.0%) 0 lost to follow-up 1 (1.3%) 2 (4.3%) patient request unrelated to adverse event 1 (1.3%) 1 (2.2%) other 1 (1.3%) 3 (6.5%) Total patients who completed study 62 (82.7%) 26 (56.5%) a Data numbers of patients (% of group total). Each reason is unique to each patient; no patient is listed more than once. min after completing her first 300-mg iron sucrose administration. Hydrocortisone was administered. Symptoms and findings resolved within 2 h. The patient elected to withdraw from the study. The investigator considered the event moderate in intensity and probably related to study drug. A second patient experienced pruritus 18 h after receiving a 400-mg dose of iron sucrose. Diphenhydramine was administered orally. Symptoms resolved within 2 d. The event was considered moderate and related to study drug. Three episodes of hypotension were recorded, two in group A, including one judged unlikely to be related and one unrelated to study drug, and one in group B. Five patients in group A had a history of intolerance to an intravenous iron product, including three who were intolerant to ferric gluconate (two with pruritic rash and one with hypotension, nausea, vomiting, and diarrhea) and two who were intolerant to iron dextran (one hypotension with back pain and one bronchospasm). The patient with leg swelling described above was one of the two patients with a history of rash after ferric gluconate. There were 11 episodes of peritonitis: Six (8.0%) in group A and five (10.9%) in group B. Of these, four episodes (two in each group) were considered serious; no episode was considered related to study drug. Similarly, there were seven episodes of catheter exit-site infection: Three (4.0%) in group A and four (8.7%) in group B. Post hoc calculations suggested that our study had sufficient power to have an 80% or better chance to detect a two-fold increase in the rate of infection (peritonitis plus exit-site infection) in group A compared with group B. During the study, 10.7% (eight of 75) of the patients in group A experienced at least one nonserious adverse event that was thought to be possibly or probably related to study drug (ADE). Among nonserious ADE reported, gastrointestinal complaints were seen most frequently (Table 3). Among the nonserious ADE was the finding of asymptomatic peripheral eosinophilia (27%) in the third week of treatment that resolved to baseline (5 to 7%) within 2 mo in a patient with a history of seasonal allergies and a failed renal transplant. The finding was rated as possibly related to intravenous iron by the investigator. Laboratory Evaluation. There were no significant changes in clinical biochemistry laboratory results, including blood ureas nitrogen, creatinine, albumin, bicarbonate, or phosphorus, at any interval in either treatment group. Discussion We report the results of a prospective, randomized, controlled trial in patients with PD-CKD that compared the anemia management efficacy of intravenous iron therapy added to ongoing ESA treatment with that of ESA therapy alone. Our results show that intravenous iron administration increases Hb without an increase in ESA dose, decreases the need for anemia intervention, and replenishes iron stores. Our trial highlights Hb-targeting bias as an inherent limitation of anemia adjuvant trials. Examination of efficacy of an adjuvant to ESA requires maintaining ESA doses constant before and after randomization to either adjuvant therapy or no-treatment control. We set the lower limit of Hb by design, and when Hb fell below that target, we removed patients from the trial to permit definitive treatment (anemia intervention). We observed an effective, albeit unspecified and off-protocol, upper Hb limit when investigators decreased ESA doses when Table 3. Reported ADE in group A safety-evaluable patients a Adverse Drug Events Group A (ESA IV Iron; n 75) Gastrointestinal 5 (6.7%) constipation 1 (1.3%) diarrhea 2 (2.7%) nausea/vomiting 2 (2.7%) abdominal pain 1 (1.3%) Pruritus and swelling 1 (1.3%) Pruritus 1 (1.3%) Discoloration at injection site 1 (1.3%) Increased peripheral eosinophil 1 (1.3%) count a Data are number of patients (% column total). All events were judged nonserious. One patient reported two gastrointestinal adverse drug events.

Clin J Am Soc Nephrol : -, Intravenous Iron in PD Patients Who Receive ESA Therapy 7 a high Hb raised safety or reimbursement concerns. The resulting Hb targeting in our anemia adjuvant trial, like dose targeting in a dialysis adequacy trial (8), introduces unavoidable bias. Control group patients in our trial were more likely to need ESA dose increases and other anemia interventions and therefore were more likely to be removed from the trial. Greater dropout rates in the no-iron control group, as seen previously in a trial of oral iron versus no-iron treatment in patients with HD-dependent CKD (9), create selective loss (censoring) of patients who are most prone to poor anemia outcomes and leaves the remaining control group patients relatively responsive to ESA and dietary iron. Iron-deficient patients without kidney disease may show increases in both Hb and TSAT despite assignment to treatment with an iron-free placebo (10), presumably because dietary iron intake in healthy individuals is sufficient to provide positive iron balance. In short, in patients who are receiving ESA therapy, Hb targeting bias minimizes differences that are achieved between iron treatment and no-iron control arms. Even with these constraints, however, intravenous iron administration proved effective in our study as an adjuvant to ESA therapy. In patients who had PD-CKD and underwent combined ESA and intravenous iron therapy, Hb rose higher, interventions were fewer, and net epoetin dose decrease was greater than in patients who were given ESA therapy alone. Whether oral iron administration is an effective adjunct to ESA therapy in PD patients is unclear. Failure of oral iron administration to sustain target iron indices in PD patients who undergo ESA therapy is common (1,11). When intravenous iron is administered to PD patients who previously had received oral iron, Hb levels rise and ESA doses either fall or remain unchanged (2,4,6,7). Although intravenous iron agents are more expensive than oral iron agents, the cost of an effective ESA adjuvant is offset by sparing of the cost of ESA. In ESAtreated patients, therefore, failure of an inexpensive adjuvant may increase the effective cost of anemia management. Although RCT have shown intravenous iron to be superior to oral iron in ESA-treated HD patients (12 14) and patients with CKD (15), RCT in PD patients are currently lacking. Our study provides needed information to guide the design, size, and duration of such a comparative trial. In HD patients, the iron status tests TSAT and ferritin are useful markers of responsiveness to a 1000-mg intravenous iron challenge (16). In the patients with PD-CKD in this study, characterized by ESA use and moderate iron status (Hb 11.5 g/dl, TSAT 25%, ferritin 500 ng/ml), these tests proved unhelpful (Figure 6). Remarkably, despite the fact that few patients met criteria for iron deficiency at baseline CKD (TSAT 20% and ferritin 100 ng/ml; Table 1), nearly 60% of patients in this study showed a 1-g/dl rise in Hb after intravenous iron administration. This success rate, coupled with the observation that the likelihood of Hb increase seemed to be independent of iron status (Figure 6), confirms that iron deficiency is not required to demonstrate intravenous iron efficacy in patients with PD-CKD (7). Convenient dosing schedules are important practical determinants of the efficacy of intravenous iron administration in the outpatient treatment setting. The maximum Food and Drug Administration approved dose for single administration is 100 mg for iron dextran over 2 min and 125 mg of ferric gluconate over 10 min. Iron sucrose is Food and Drug Administration approved for administration of 100 mg over 2 to 5 min, 200 mg over 2 to 5 min, 300 mg over 1.5 h, or 400 mg over 2.5 h. Hypotension, presumably related to bioactive iron release, is a concern whenever intravenous iron is administered too much, too fast (17). Although two episodes of hypotension were recorded in patients who were assigned to group A, neither was considered either serious or likely to have been related to intravenous iron. Iron sucrose has been administered as 1000 mg in two 500-mg doses 1 to 2 wk apart, but the described infusion times of 3.5 to 6 h for each dose may prove inconvenient for many (4,7,18). We also observed two episodes of pruritus after 300- and 400-mg infusions, respectively. Whether these reactions were due to hypersensitivity or were related to rate and total dose of infusion is unclear. Hypersensitivity reactions to nondextran iron agents are less common and less severe than those seen after iron dextran (19). Because intravenous iron agents have been shown to augment the growth of pathogenic bacteria in vitro (20), we recorded infection rates in patients through 30 d after trial completion or discontinuation. In keeping with previous evidence in patients with PD-CKD (21), we found the rate of infection to be no higher among patients who received intravenous iron than among those who did not. These findings held true for rates of all peritonitis, serious peritonitis, and catheter exit-site infection. We conclude that a 1000-mg intravenous iron challenge poses no discernible infectious risk for patients with PD-CKD. Conclusion Results of the first RCT to assess intravenous iron in patients with anemia that is associated with PD-CKD confirm that intravenous iron administration can be an effective adjuvant to ESA therapy, even when baseline iron status tests do not suggest iron deficiency. Moreover, administration of iron sucrose as 300 mg over 1.5 h or 400 mg over 2.5 h is well tolerated and provides a practical approach to completing iron repletion expeditiously in the outpatient treatment center. Acknowledgment This study was supported by American Regent, Inc. References 1. Ahsan N: Intravenous infusion of total dose iron is superior to oral iron in treatment of anemia in peritoneal dialysis patients: A single center comparative study. JAmSoc Nephrol 9: 664 668, 1998 2. Johnson DW, Herzig KA, Gissane R, Campbell SB, Hawley CM, Isbel NM: A prospective crossover trial comparing intermittent intravenous and continuous oral iron supplements in peritoneal dialysis patients. Nephrol Dial Transplant 16: 1879 1884, 2001 3. Ahsan N, Groff JA, Waybill MA: Efficacy of bolus intravenous iron dextran treatment in peritoneal dialysis patients

8 Clinical Journal of the American Society of Nephrology Clin J Am Soc Nephrol : -, receiving recombinant human erythropoietin. Adv Perit Dial 12: 161 166, 1996 4. Prakash S, Walele A, Dimkovic N, Bargman J, Vas S, Oreopoulos D: Experience with a large dose (500 mg) of intravenous iron dextran and iron saccharate in peritoneal dialysis patients. Perit Dial Int 21: 290 295, 2001 5. Sloand JA, Shelly MA, Erenstone AL, Schiff MJ, Talley TE, Dhakal MP: Safety and efficacy of total dose iron dextran administration in patients on home renal replacement therapies. Perit Dial Int 18: 522 527, 1998 6. Richardson D, Bartlett C, Jolly H, Will EJ: Intravenous iron for CAPD populations: Proactive or reactive strategies? Nephrol Dial Transplant 16: 115 119, 2001 7. Domrongkitchaiporn S, Jirakranont B, Atamasrikul K, Ungkanont A, Bunyaratvej A: Indices of iron status in continuous ambulatory peritoneal dialysis patients. Am J Kidney Dis 34: 29 35, 1999 8. Greene T, Daugirdas J, Depner T, Allon M, Beck G, Chumlea C, Delmez J, Gotch F, Kusek JW, Levin N, Owen W, Schulman G, Star R, Toto R, Eknoyan G; Hemodialysis Study Group: Association of achieved dialysis dose with mortality in the Hemodialysis study: An example of dose-targeting bias. J Am Soc Nephrol 16: 3371 3380, 2005 9. Markowitz GS, Kahn GA, Feingold RE, Coco M, Lynn RI: An evaluation of the effectiveness of oral iron therapy in hemodialysis patients receiving recombinant human erythropoietin. Clin Nephrol 48: 34 40, 1997 10. Krafft A, Perewusnyk G, Hanseler E, Quack K, Huch R, Breymann C: Effect of postpartum iron supplementation on red cell and iron parameters in non-anaemic iron-deficient women: A randomised placebo-controlled study. BJOG 112: 445 450, 2005 11. Nissenson AR, Korbet S, Faber M, Burkart J, Gentile D, Hamburger R, Mattern W, Schreiber M, Swartz R, Van Stone J: Multicenter trial of erythropoietin in patients on peritoneal dialysis. J Am Soc Nephrol 5: 1517 1529, 1995 12. Fishbane S, Lynn RI: The efficacy of iron dextran for the treatment of iron deficiency in hemodialysis patients. Clin Nephrol 44: 238 240, 1995 13. Macdougall IC, Tucker B, Thompson J, Tomson CR, Baker LR, Raine AE: A randomized controlled study of iron supplementation in patients treated with erythropoietin. Kidney Int 50: 1694 1699, 1996 14. Fudin R, Jaichenko J, Shostak A, Bennett M, Gotloib L: Correction of uremic iron deficiency anemia in hemodialyzed patients: A prospective study. Nephron 79: 299 305, 1998 15. Van Wyck DB, Roppolo M, Martinez CO, Mazey RM, McMurray S; for the United States Iron Sucrose (Venofer) Clinical Trials Group: A randomized, controlled trial comparing IV iron sucrose to oral iron in anemic patients with nondialysis-dependent CKD. Kidney Int 68: 2846 2856, 2005 16. IV. NKF-K/DOQI clinical practice guidelines for anemia of chronic kidney disease: Update 2000. Am J Kidney Dis 37: S182 S238, 2001 17. Van Wyck DB: Labile iron: Manifestations and clinical implications. J Am Soc Nephrol 15[Suppl 2]: S107 S111, 2004 18. Agarwal HK, Tziviskou E, Bellizzi V, Khandelwal M, Moupas L, Bargman JM, Jassal SV, Oreopoulos DG: Prolonged administration over 6 hours of large doses of intravenous iron saccharate (500 mg) prevents severe reactions in peritoneal dialysis patients. Perit Dial Int 22: 636 637, 2002 19. Bailie GR, Clark JA, Lane CE, Lane PL: Hypersensitivity reactions and deaths associated with intravenous iron preparations. Nephrol Dial Transplant 20: 1443 1449, 2005 20. Parkkinen J, von Bonsdorff L, Peltonen S, Gronhagen- Riska C, Rosenlof K: Catalytically active iron and bacterial growth in serum of haemodialysis patients after i.v. ironsaccharate administration. Nephrol Dial Transplant 15: 1827 1834, 2000 21. Allen JR, Troidle LK, Juergensen PH, Kliger AS, Finkelstein FO: Incidence of peritonitis in chronic peritoneal dialysis patients infused with intravenous iron dextran. Perit Dial Int 20: 674 678, 2000