Optimizing the management of renal anemia: challenges and new opportunities

Optimizing the management of renal anemia: challenges and new opportunities

review http://www.kidney-international.org & 2008 International Society of Nephrology Optimizing the management of renal anemia: challenges and new ...

117KB Sizes 0 Downloads 53 Views

review

http://www.kidney-international.org & 2008 International Society of Nephrology

Optimizing the management of renal anemia: challenges and new opportunities Francesco Locatelli1 and Lucia Del Vecchio1 1

Department of Nephrology, Dialysis and Renal Transplant, Ospedale A. Manzoni, Lecco, Italy

Erythropoiesis stimulating agents (ESAs) are the main tool to achieve anemia correction in CKD patients. At present six different ESAs are available: epoetin alpha, epoetin beta, epoetin omega, epoetin delta, darbepoetin alpha, and very recently CERA. From one side the patent of older ESAs have expired, and biosimilars (for the moment only of epoetin alpha) have been approved for use in Europe by the European Medicines Agency. However, a number of issues about bioequivalence and how to test it are still to be solved completely. In the mean time pharmaceutical research has kept on working, developing new ESAs and alternative strategies for stimulating erythropoiesis. In this review we present and discuss these points.

Anemia, resulting primarily from insufficient production of erythropoietin (EPO) to support erythropoiesis, is a common complication of chronic kidney disease (CKD). Approximately 50% of patients with CKD stages 3–5 have anemia.1 An even greater proportion of patients starting dialysis are anemic.2 Anemia may significantly impair quality of life, increase cardiovascular risk, and reduce long-term survival, when left untreated. Earlier, treatment options were essentially limited to blood transfusions; however, since the late 1980s the availability of recombinant human EPO (rHuEPO) has revolutionized the management of anemia in patients with CKD. Today, erythropoiesis-stimulating agents (ESAs) are the main tools to achieve anemia correction in CKD patients.

Kidney International (2008) 74 (Suppl 111), S33–S37; doi:10.1038/ki.2008.525 KEYWORDS: anemia; chronic kidney disease; erythropoietin; erythropoiesisstimulating agents; hematide; continuous erythropoiesis receptor activator

Correspondence: Francesco Locatelli, Department of Nephrology, Dialysis and Renal Transplant, Ospedale A. Manzoni, Via dell’Eremo 9, 23900 Lecco, Italy. E-mail: [email protected] Kidney International (2008) 74 (Suppl 111), S33–S37

TYPES OF HUMAN RECOMBINANT EPO

The gene encoding for HuEPO was cloned in 1985.3 This opened the way to the synthesis of rHuEPO.4,5 This molecule has been proven effective and relatively safe in correcting anemia and maintaining stable hemoglobin (Hb) levels in CKD patients. Currently, four different types of rHuEPO are available in the market: epoetin alpha, epoetin beta, epoetin omega, and epoetin delta. Both epoetin alpha and beta are synthesized in Chinese hamster ovary cells and share the same amino acid sequence as endogenous EPO, but differences in the manufacturing process between the two glycoproteins reflect into differences in their carbohydrate moieties.6 Epoetin omega is synthesized in baby hamster kidney cells.7 Differing from epoetin alpha and beta, only 60% of epoetin omega is O-glycosylated at the serine residue, and a higher proportion of carbohydrate residues are tetrasialylated when compared with endogenous EPO, as has been reported for the other epoetins.8 Epoetin delta shares the same amino acid sequence as endogenous EPO, but is synthesized in human cells.9,10 This process may circumvents problems arising from species-dependent differences in protein-folding or post-translational modification. Despite these differences in the glycosylation pattern, all rHuEPO have similar half-life (6–8 h intravenously and 19–24 h subcutaneously), pharmacodynamic and pharmacokinetic characteristics. BIOSIMILARS

In December 2004, the patent of epoetin alpha expired in Europe, followed by epoetin beta in 2005 in many European S33

countries. This has opened the way to biosimilars. Currently, two biosimilars of epoetin alpha, HX575 (Rentschler Biotechnologie, Laupheim, Germany) and Epoetin zeta (Silapo by STADA Arzneimittel AG, Bad Vilbel, Germany and Retacrit by Hospira Enterprises B.V., Hoofddorp, the Netherland), received full marketing authorization valid throughout the European Union. HX575, which is already available in some European countries, is marketed by three different companies (‘Binocrit’ from Sandoz GmbH, Holzkirchen, Germany, ‘Epoetin alfa Hexal’ from Hexal Biotech Forschungs GmbH, Holzkirchen, Germany and ‘Abseamed’ from Medice Arzneimittel Pu¨tter GmbH & Co., Iserlohn, Germany). Both drugs are not inferior to epoetin alpha in maintaining target Hb concentrations; they can be administered only intravenously in CKD patients. Unfortunately, the fulfillment of the European Medicines Agency requirements for approval does not solve completely the issue of what bioequivalence really means and how it has to be tested, as quality assurance assays for biopharmaceuticals are generally less sensitive and precise than those for small molecules, and many of them still require development and standardization.11 THE NEW GENERATION OF ESAs

Even though it is very effective in correcting anemia, rHuEPO cannot be considered as the ideal ESA. It requires a relatively frequent administration and this has been claimed as a possible reason of high Hb variability. Moreover, as its oral bioavailability is insufficient, it can be administered only by the subcutaneous or intravenous route, the latter requiring higher doses than the subcutaneous one. In the setting of hemodialysis, where the intravenous route is more convenient, physicians have to balance increased costs because of extra dose needs with patient discomfort and risk of bloodrelated infections. Another important limitation of rHuEPO is that it is unstable at room temperature. A strict cold chain is thus necessary, starting from the manufacturer till the moment of administration; an inadequate handling of the drug may increase the risk of immunization in patients selfadministering the drug at home. Finally, the synthesis of rHuEPO, as that of other biomolecules, is complex and thus expensive. Altogether, these points have urged pharmacological research in developing new ESAs with improved characteristics. LONG-ACTING EPOs

Darbepoetin alpha is an ESA with prolonged half-life. As in epoetin alpha and beta, it is produced in Chinese hamster ovary cells but it differs from EPO in the amino acid sequence at five positions allowing the adding of two extra N-linked carbohydrate chains.12 These molecular changes result in longer circulating half-life (B25 h when given intravenously and B48 h by the subcutaneous route) and in a 4.3-fold lower relative affinity for the EPO receptor. Given its longer half-life, darbepoetin alpha can be administered less often than rHuEPO.13–16 Differing from rHuEPO, dose requirements are S34

F Locatelli and L Del Vecchio: Optimizing renal anemia management

independent of the administration route.14,17 According to the label, the drug can remain at room temperature (up to 251C) for a maximum single period of 7 days. Continuous erythropoiesis receptor activator (CERA) is another new ESA recently become available that is obtained by adding a large water-soluble polyethylene glycol moiety to the EPO beta molecule. This agent has a much higher molecular weight than that of EPO (B60,000 Da vs 34,000 Da) and a longer half-life (B130 h when administered either intravenously or subcutaneously). Moreover, it has a reduced binding affinity for the EPO receptor, which is B45-fold lower than that of epoetin beta, mainly because of a much slower association rate.18 Given these binding properties, the molecule activates the EPO receptor but is not internalized into the cell, remaining are available for binding to other EPO receptors. Results of clinical trials indicate that CERA is effective in maintaining Hb levels after switching from rHuEPO therapy (one to three times a week) or darbepoetin alpha (once a week or once every other week) when administered once every 4 weeks;19,20 and in correcting anemia in EPO-naive patients when administered once in every 2 weeks.21 Since the publication in 2006 of two trials aimed at testing the effect of complete anemia correction on cardiovascular status and survival in CKD patients not on dialysis,22,23 a lively debate is going on in the nephrological community about the fear of complications consequent to anemia overcorrection. In this perspective, it is of interest that the drug achieves the Hb target more smoothly than other ESAs, reducing the risk of overshooting (Figure 1).21 Moreover, despite a longer half-life than that of other ESAs, when the drug is halted because Hb levels exceed the target, the rate of decrease in Hb levels is similar to that observed with short-acting ESAs. This suggests that the half-life of erythrocytes is more important than that of the drug (Figure 2). As for darbepoetin alpha, the dose does not need to be modified according to the adminisCERA 1x/2wk Mean (s.d.) Hb (g per 100 ml)

review

Darbepoetin alfa 1x/wk

16 15

rHuEPO

14 13 12 11

10 9 8 7 Months Weeks

BL

1

2

3

4

5

6

4

8

12

16

20

24

Final visit

Figure 1 | Hb concentrations during correction phase with CERA, rHuEPO, and darbepoetin alpha in EPO-naive CKD patients not on dialysis. The observed raise in Hb concentration is smothering with CERA than with other ESAs. Data are obtained from reference Macdougall et al.21 and from reference Locatelli et al.13 CERA, continuous erythropoiesis receptor activator; CKD, chronic kidney disease; Hb, hemoglobin; rHuEPO, recombinant human erythropoietin. Kidney International (2008) 74 (Suppl 111), S33–S37

review

F Locatelli and L Del Vecchio: Optimizing renal anemia management

CERA QM Darbepoetin Q2W rHuEPO TIW

15

Hb (g per 100 ml)

14 13 12 11 10 –6.0

–3.5

–1.0

1.5

4.0

6.5

9.0

11.5

Weeks after drug withdrawal

Figure 2 | Hb decrease rate after drug withdrawal in patients exceeding Hb levels of 14 g per 100 ml with different ESAs. The rate of decrease is influenced by erythrocyte- and not by drug half-life. Data are obtained from reference Locatelli et al.13 and reference Heifets et al.49 CERA, continuous erythropoiesis receptor activator; ESAs, Erythropoiesis-stimulating agents; Hb, hemoglobin.

tration route. The drug can remain at room temperature for a maximum single period of nearly one month. FUSION PROTEINS AND SYNTHETIC EPO

Beyond increasing the carbohydrate content of EPO, various strategies have been pursued to increase the yield of EPO produced by gene transfer and to enhance its intrinsic activity. Erythropoietin binds to the EPO receptor through two binding domains, one with high affinity for the receptor and the other with low affinity; the latter is required for the activation of the receptor. On the basis of the hypothesis that the presence of two molecules of EPO might bring together two high-affinity binding sites and facilitate binding to the EPO receptor, one approach is the production of a molecule consisting of two EPOs linked by small flexible peptides.24,25 Experimental data indicate that EPO dimers induce a much higher hematocrit increase than the EPO monomer without differences in pharmacokinetics.25 It has been shown that fusing the carboxyl-terminal peptide of human chorionic gonadotropin beta-subunit (human chorionic gonadotropin beta) to a number of biologically active proteins increased in vivo potency and circulatory half-life of the proteins containing this peptide without affecting assembly, secretion, receptor binding affinity, or in vitro bioactivity. Recently, Fares et al.26 obtained a fusion protein by adding the carboxyl-terminal peptide of human chorionic gonadotropin beta to HuEPO-coding sequence. This chimeric protein had both enhanced in vivo potency and half-life compared with wild-type EPO in mice. Another approach for enhancing serum half-life includes increasing the molecular weight of EPO by fusion to the Fc part of an antibody.27,28 Interestingly, this approach has been used to create an Fc fusion protein that can be administered by aerosol inhalation.29 Attempts have also been made to synthesize artificially a glycoprotein similar to EPO, called synthetic erythropoiesis Kidney International (2008) 74 (Suppl 111), S33–S37

protein. This is a 51-kDa protein-polymer consisting of 166 amino acids with two other polymer moieties attached covalently.30 In cell and animal assays for erythropoiesis, synthetic erythropoiesis protein displayed potent biological activity and had significantly prolonged duration of action in vivo.31 Even though it is effective in correcting anemia, all these EPO-modified proteins raise some concerns about immunogenicity.32 NON-EPO-DERIVED EPO RECEPTOR AGONISTS: HEMATIDE

Hematide is a pegylated synthetic dimeric peptide with no sequence homology to EPO. In CKD patients not on dialysis, it has an intermediate mean half-life amid darbepoetin alpha and CERA (B59 and B70 h by the intravenous and subcutaneous route, respectively) and longer half-life than that measured in healthy individuals; this is consistent with the hypothesis that hematide is cleared, at least partially, through the kidney.33 Phase II trials showed that the drug given either intravenously or subcutaneously was effective in both the correction and maintenance phase of anemia treatment at dose ranges between 0.025 and 0.05 mg/kg; phase III trials are ongoing. Even though antibodies against hematide have been found in patients receiving the drug, they do not cross react with EPO.34 For this reason, hematide has been proposed as a possible treatment of anemia in pure red cell aplasia.34,35 Potential advantage of this drug is that its manufacturing process is simpler and cheaper than that of current ESAs. Recently, in the attempt to create other non-EPO-derived EPO receptor agonists, two sequences of EPO mimetic peptide-1, which is a 20-amino acid peptide with weak EPOlike bioactivity, were inserted on a human IgG4 Fc framework. The obtained molecule, called CNTO 530, was shown to bind selectively the EPO receptor, to mediate EPO receptor signaling, and to support proliferation of EPO-dependent cells.36 In vivo it stimulated erythropoiesis, providing a longlived increase in Hb in mice. OTHER STRATEGIES TO INCREASE ERYTHROPOIESIS

The inhibition or activation of transcription factors modulating EPO gene expression may be an alternative for stimulating erythropoiesis. The hypoxia-inducible transcription factors (HIFs) are central components in the cellular responses to hypoxia; under normoxic conditions, EPO gene expression is suppressed as a result of HIF inactivation because of O(2)dependent enzymatic hydroxylation and subsequent degradation of their alpha-subunit. 2-oxoglutarate analogs, which prevent HIF alpha hydroxylation, have emerged as promising tools for the stimulation of erythropoiesis and angiogenesis (the so called HIF stabilizers). Experimental data indicate that these agents stimulate erythropoiesis in vivo.37 These agents can be administered by the oral route. Unfortunately, the development of the first candidate molecule of this class, FG-2216, was halted after the occurrence of a case of fatal S35

review

hepatic necrosis. Another concern related to HIF stabilizers is related to the fact that the HIF system promotes the transcription of many other genes; careful evaluation of other effects than erythropoiesis stimulation seems mandatory. GATA genes are an evolutionarily conserved family, which encode a group of important transcription factors involved in the regulation of diverse processes including the development of the heart, hematopoietic system, and sex gonads. Some of these factors also regulate EPO gene transcription.38,39 Oral administration of a GATA-specific inhibitor, K-7174, was found effective in reversing anemia induced by IL-1 beta or TNF-a in mice.40 Another possible strategy to increase erythropoiesis is the inhibition of the hematopoietic cell phosphatase. This protein, also known as src homology domain 2-containing tyrosine phosphatase 1, binds and activates the negative regulatory domain of the EPO receptor, inhibiting transduction inside the cell.41 Interestingly, a defective expression of hematopoietic cell phosphatase was found in patients with polycythemia vera.42 In contrast, hematopoietic cell phosphatase expression is increased in CD34 þ cells from patients with EPO resistance; the adding of a src homology domain 2-containing tyrosine phosphatase 1 antisense oligonucleotide suppressed src homology domain 2-containing tyrosine phosphatase 1 protein expression and partial recovered BFU-E colonies.43 Hematopoietic cell phosphatase inhibitors may be thus a novel target molecule to treat renal anemia and/or sensitize patients to EPO action. Finally, EPO gene therapy has been proposed for the management of anemic patients with CKD. A number of different techniques have been tried, such as the delivering of naked plasmid DNA into skeletal muscles,44,45 the subcutaneous implantation of autologous bone marrow stromal cells genetically engineered to secrete EPO,46 viral transfection,47 and the use of artificial human chromosome.48 EPO gene therapy has the theoretical advantage of releasing small but continuous amount of EPO into the circulation, but has still a number of problems, such as immunogenicity and the difficulty to exactly tune the exact amount of EPO needed to correct anemia and maintaining a level of expression that sufficiently promotes erythropoiesis in the long term.

F Locatelli and L Del Vecchio: Optimizing renal anemia management

DISCLOSURE

Francesco Locatelli has received consulting fees from Affimax, Roche, Amgen, and Shire. Francesco Locatelli has also received lecture fees from Roche, Amgen, and Shire. Lucia Del Vecchio has declared no financial interests. REFERENCES 1.

2.

3. 4.

5.

6.

7.

8.

9.

10.

11. 12.

13.

14.

15.

16.

17.

CONCLUSIONS

Currently, the treatment of anemia with rHuEPO is quite expensive, given that its synthesis, as that of other biomolecules, is complex. New ESAs are, or will be, more expensive than rHuEPO, because of the high costs of pharmacological research, not yet covered by years of selling, but offer improved pharmacological characteristics that may help in the management of anemia in CKD patients. The synthesis of simpler molecules that is not related to ESA structure or the development of alternative strategies of erythropoiesis stimulation may lead to the creation of cheaper agents that would revolutionize the market. S36

18.

19.

20.

Mcclellan WM, Aronoff SL, Bolton WK. The prevalence of anaemia in patients with chronic kidney disease. Curr Med Res Opin 2004; 20: 1501–1510. Locatelli F, Pisoni RL, Combe C et al. Anaemia in haemodialysis patients of five European countries: association with morbidity and mortality in the Dialysis Outcomes and Practice Patterns Study DOPPS). Nephrol Dial Transplant 2004; 19: 121–132. Lin FK, Suggs S, Lin CH et al. Cloning and expression of the human erythropoietin gene. Proc Natl Acad Sci USA 1985; 82: 7580–7584. Winearls CG, Oliver DO, Pippard MJ et al. Effect of human erythropoietin derived from recombinant DNA on the anaemia of patients maintained by chronic haemodialysis. Lancet 1986; 2: 1175–1178. 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. Storring PL, Tiplady RJ, Gaines Das RE et al. Epoetin alfa and beta differ in their erythropoietin isoform compositions and biological properties. Br J Haematol 1998; 100: 79–89. Powell JS, Berkner KL, Lebo RV et al. Human erythropoietin gene: high level expression in stably transfected mammalian cells and chromosome localization. Proc Natl Acad Sci USA 1986; 83: 6465–6469. Nimtz M, Martin W, Wray V et al. Structures of sialylated oligosaccharides of human erythropoietin expressed in recombinant BHK-21 cells. Eur J Biochem 1993; 213: 39–56. Deicher R, Horl WH. Differentiating factors between erythropoiesisstimulating agents: a guide to selection for anaemia of chronic kidney disease. Drugs 2004; 64: 499–509. Kwan JT, Pratt RD, The Epoetin Delta Study Group. Epoetin delta, erythropoietin produced in a human cell line, in the management of anaemia in predialysis chronic kidney disease patients. Curr Med Res Opin 2007; 23: 307–311. Locatelli F, Roger S. Comparative testing and pharmacovigilance of biosimilars. Nephrol Dial Transplant 2006; 21(Suppl 5): v13–v16. Egrie JC, Browne KJ. Development and characterisation of novel erythropoiesis stimulating protein (NESP). Nephrol Dial Transplant 2001; 16(Suppl 3): 3–13. Locatelli F, Olivares J, Walker R et al. European/Australian NESP 980202 Study Group. Novel erythropoiesis stimulating protein for treatment of anemia in chronic renal insufficiency. Kidney Int 2001; 60: 741–747. Locatelli F, Canaud B, Giacardy F et al. Treatment of anaemia in dialysis patients with unit dosing of darbepoetin alfa at a reduced dose frequency relative to recombinant human erythropoietin (rHuEpo). Nephrol Dial Transplant 2003; 18: 362–369. Vanrenterghem Y, Barany P, Mann JF et al. European/Australian NESP 970200 Study Group. Randomized trial of darbepoetin alfa for treatment of renal anemia at a reduced dose frequency compared with rHuEPO in dialysis patients. Kidney Int 2002; 62: 2167–2175. Mann J, Kessler M, Villa G et al. Darbepoetin alfa once every 2 weeks for treatment of anemia in dialysis patients: a combined analysis of eight multicenter trials. Clin Nephrol 2007; 67: 140–148. Aarup M, Bryndum J, Dieperink H et al. Clinical implications of converting stable haemodialysis patients from subcutaneous to intravenous administration of darbepoetin alfa. Nephrol Dial Transplant 2006; 21: 1312–1316. Brandt M, Lazendo¨rfer M, Frische J et al. Different receptor binding activity of C.E.R.A. (continuous erythropoietin receptor activator) compared with epoetin beta determined by surface plasmon resonance and competition assay on UT-7 cells. Nephrol Dial Transplant 2006; 21(Suppl 4): iv9 (Abstract SO018). Locatelli F, Villa G, De Francisco ALM et al. Effect of a continuous erythropoietin receptor activator (C.E.R.A.) on stable haemoglobin in patients with CKD on dialysis. Curr Med Res Opin 2007; 23: 969–979. Levin NW, Fishbane S, Can˜edo FV et al. MAXIMA study investigators: intravenous methoxy polyethylene glycol-epoetin beta for haemoglobin control in patients with chronic kidney disease who are on dialysis: a randomised non-inferiority trial (MAXIMA). Lancet 2007; 370: 1415–1421. Kidney International (2008) 74 (Suppl 111), S33–S37

review

F Locatelli and L Del Vecchio: Optimizing renal anemia management

21.

22.

23.

24.

25.

26.

27.

28.

29.

30.

31.

32.

33.

34.

35.

Macdougall IC, Walker R, Provenzano R et al. C.E.R.A. (continuous erythropoietin receptor activator) administered at extended intervals corrects anemia and maintains stable Hb levels in patients with CKD not on dialysis. J Am Soc Nephrol 2006; 17: 619A (Abstract SA-PO208). Drueke TB, Locatelli F, Clyne N, et al., CREATE Investigators. Normalization of hemoglobin level in patients with chronic kidney disease and anemia. N Engl J Med 2006; 355: 2071–2084. Singh AK, Szczech L, Tang KL, et al., CHOIR Investigators. Correction of anemia with epoetin alfa in chronic kidney disease. N Engl J Med 2006; 355: 2085–2098. Sytkowski AJ, Lunn ED, Davis KL et al. Human erythropoietin dimers with markedly enhanced in vivo activity. Proc Natl Acad Sci USA 1998; 95: 1184–1188. Dalle B, Henri A, Rouyer-Fessard P et al. Dimeric erythropoietin fusion protein with enhanced erythropoietic activity in vitro and in vivo. Blood 2001; 97: 3776–3782. Fares F, Ganem S, Hajouj T et al. Development of a long-acting erythropoietin by fusing the carboxyl-terminal peptide of human chorionic gonadotropin beta-subunit to the coding sequence of human erythropoietin. Endocrinology 2007; 148: 5081–5087. Schriebl K, Trummer E, Lattenmayer C et al. Biochemical characterization of rhEpo-Fc fusion protein expressed in CHO cells. Protein Expr Purif 2006; 49: 265–275. Way JC, Lauder S, Brunkhorst B et al. Improvement of Fc-erythropoietin structure and pharmacokinetics by modification at a disulfide bond. Protein Eng Des Sel 2005; 18: 111–118. Dumont JA, Bitonti AJ, Clark D et al. Delivery of an erythropoietin-Fc fusion protein by inhalation in humans through an immunoglobulin transport pathway. J Aerosol Med 2005; 18: 294–303. Kochendoerfer GG, Chen SY, Mao F et al. Design and chemical synthesis of a homogeneous polymer-modified erythropoiesis protein. Science 2003; 299: 884–887. Chen SY, Cressman S, Mao F et al. Synthetic erythropoietic proteins: tuning biological performance by site-specific polymer attachment. Chem Biol 2005; 12: 371–383. Coscarella A, Liddi R, Di Loreto M et al. The rhGM-CSF-EPO hybrid protein MEN 11300 induces anti-EPO antibodies and severe anaemia in rhesus monkeys. Cytokine 1998; 10: 964–969. Fan Q, Leuther KK, Holmes CP et al. Preclinical evaluation of Hematide, a novel erythropoiesis stimulating agent, for the treatment of anemia. Exp Hematol 2006; 34: 1303–1311. Woodburn KW, Fan Q, Winslow S et al. Hematide is immunologically distinct from erythropoietin and corrects anemia induced by antierythropoietin antibodies in a rat pure red cell aplasia model. Exp Hematol 2007; 35: 1201–1208. NCT00314795 registered on the clinicaltrial.gov website.

Kidney International (2008) 74 (Suppl 111), S33–S37

36.

37.

38. 39.

40.

41.

42.

43.

44.

45.

46.

47.

48.

49.

Bugelski PJ, Capocasale RJ, Makropoulos D et al. CNTO 530: molecular pharmacology in human UT-7EPO cells and pharmacokinetics and pharmacodynamics in mice. J Biotechnol 2008; 134: 171–180. Safran M, Kim WY, O’Connell F et al. Mouse model for noninvasive imaging of HIF prolyl hydroxylase activity: assessment of an oral agent that stimulates erythropoietin production. Proc Natl Acad Sci USA 2006; 103: 105–110. Dame C, Sola MC, Lim K-C et al. Hepatic erythropoietin gene regulation by GATA-4. J Biol Chem 2004; 279: 2955–2961. Imagawa S, Yamamoto M, Miura Y. Negative regulation of the erythropoietin gene expression by the GATA transcription factors. Blood 1997; 89: 1430–1439. Nakano Y, Imagawa S, Matsumoto K et al. Oral administration of K-11706 inhibits GATA binding activity, enhances hypoxia-inducible factor 1 binding activity, and restores indicators in an in vivo mouse model of anaemia of chronic disease. Blood 2004; 104: 4300–4307. Klingmuller U, Lorenz U, Cantley LC et al. Specific recruitment of SH-PTP1 to the erythropoietin receptor causes inactivation of JAK2 and termination of proliferative signals. Cell 1995; 80: 729–738. Wickrema A, Chen F, Namin F et al. Defective expression of the SHP-1 phosphatase in polycythemia vera. Exp Hematol 1999; 27: 1124–1132. Akagi S, Ichikawa H, Okada T et al. The critical role of SRC homology domain 2-containing tyrosine phosphatase-1 in recombinant human erythropoietin hyporesponsive anemia in chronic hemodialysis patients. J Am Soc Nephrol 2004; 15: 3215–3224. Sebestye´n MG, Hegge JO, Noble MA et al. Progress toward a nonviral gene therapy protocol for the treatment of anemia. Hum Gene Ther 2007; 18: 269–285. Fattori E, Cappelletti M, Zampaglione I et al. Gene electro-transfer on an improved erythropoietin plasmid in mice and non-human primates. J Gene Med 2005; 7: 228–236. Eliopoulos N, Gagnon RF, Francois M et al. Erythropoietin delivery by genetically engineered bone marrow stromal cells for correction of anemia in mice with chronic renal failure. J Am Soc Nephrol 2006; 17: 1576–1584. Oh TK, Quan GH, Kim HY et al. Correction of anemia in uremic rats by intramuscular injection of lentivirus carrying an erythropoietin gene. Am J Nephrol 2006; 26: 326–334. Kakeda M, Hiratsuka M, Nagata K et al. Human artificial chromosome (HAC) vector provides long-term therapeutic transgene expression in normal human primary fibroblasts. Gene Ther 2005; 12: 852–856. Heifets M, Dougherty FC, Beyer U et al. on behalf of the PROTOS, MAXIMA, STRIATA and RUBRA study investigators. Hemoglobin (Hb) decline after withholding CERA. Administration is not influenced by the long half life of CERA. Abstract presented at the World Congress of Nephrology, 2007.

S37