Pink Sheet is part of Pharma Intelligence UK Limited

This site is operated by Pharma Intelligence UK Limited, a company registered in England and Wales with company number 13787459 whose registered office is 5 Howick Place, London SW1P 1WG. The Pharma Intelligence group is owned by Caerus Topco S.à r.l. and all copyright resides with the group.

This copy is for your personal, non-commercial use. For high-quality copies or electronic reprints for distribution to colleagues or customers, please call +44 (0) 20 3377 3183

Printed By

UsernamePublicRestriction

EMA reflection paper on generic nanoparticle iron products: a case of bias by omission?

This article was originally published in SRA

Nanoparticle iron (NPI) products are used as diagnostic MRI contrast agents and as therapies for iron-deficiency anaemia. David Snodin argues that a European Medicines Agency working party should reconsider its position in its reflection paper that recommends comparative non-clinical studies for generic NPI products.

A reflection paper prepared for the European Medicines Agency's scientific committee, the CHMP, appears, at a superficial level, to present a plausible rationale for requiring comparative non-clinical studies on generic parenteral nanoparticle iron (NPI) products1. However, a more detailed and critical assessment of the data in the document, and of important evidence omitted, suggests otherwise.

The one-sided and cursory narrative presented in the paper raises fundamental issues in relation to fairness and balance in regulatory decision making and animal welfare.

NPI products are many and varied, falling into two broad categories: diagnostics (MRI contrast agents)2-5 and treatments for iron-deficiency anaemia6. At least seven of the latter category are approved – or close to being approved – in the EU/US7 and generic versions are becoming available in some cases, particularly for iron sucrose (originator trade name Venofer)8-10.

NPI preparations for treating iron-deficiency anaemia contain iron oxide/hydroxide nanoparticles surrounded by a carbohydrate shell that isolates bioactive iron from plasma components until the complex enters macrophages of the reticuloendothelial system mainly in the liver, spleen and bone marrow. Breakdown of the complex within macrophages slowly releases iron that enters the intracellular pool (eg as ferritin) or the iron is transported via plasma transferrin to erythroid precursor cells for incorporation into haemoglobin11. Some NPIs contain small amounts of free and/or “labile” iron that may be incorporated directly into transferrin12, 13.

Reflection papers are issued by the EMA in cases where it is too premature to develop a full guideline. The reflection paper in question, prepared by the CHMP’s Safety Working Party (SWP), states that physicochemical characterisation and human pharmacokinetic data are insufficient to evaluate the safety and efficacy of generic NPIs, citing published evidence on tissue distribution in vitro for different NPIs14 and on different toxicological profiles (in vivo) for several iron sucrose generic NPIs15.

The cited tissue distribution studies used cultured cells and the authors emphasise that their results do not reflect the in vivo situation. In addition, iron oxide nanoparticles (? 100nm) have been shown to be no more toxic in vitro than micrometer particles16. Published results of comparative rat toxicity studies, sponsored by the originator company, on Venofer and three different generic iron sucrose products (Generis, Ferriv and Hematin) appear to indicate potential differences in relation to markers of oxidative stress17-19. However, these findings should be interpreted with great caution for a number of reasons: all of the generic iron sucrose products failed to meet the US Pharmacopeia (USP) specification for iron sucrose injection for at least two parameters20; the dose used (40 mg/kg IV bolus) was at least ten times the maximum recommended clinical dose; healthy iron-replete rats were employed (thus exaggerating any iron-overload toxicity); and the rat studies appeared to be non-compliant with good laboratory practice, none of which was mentioned by the SWP.

One would have expected the SWP to be extra vigilant in its review of sponsored studies, possibly to the extent of undertaking a raw data audit. A recent report21 suggests that iron-overload phenomena found in young rats treated with supratherapeutic doses of iron sucrose are unlikely to be representative of effects in anaemic patients. A further sponsored study compared the clinical safety and efficacy of Venofer and a generic iron sucrose (Fer Mylan)22. The latter product received special approval in France23, 24. The study results showed that the generic iron sucrose may be less effective than the originator product (in terms of the 12.6% increase in the dose of Fer Mylan required to produce the same haemoglobin level) but no differences in clinical safety were observed. However, the extent of compliance of the generic iron sucrose with USP requirements is unknown and potential confounding factors, such as plasma ascorbate status, which has been shown to be positively correlated with increased iron bioavailability from iron sucrose25, and time-related changes in iron excretion, were not evaluated.

The possibility of toxicity mediated by oxidative stress and formation of reactive oxygen species (ROS) is a long-standing concern for iron-containing pharmaceuticals in general and, in particular, for parenteral NPI products. Originator iron sucrose has been associated with a variety of potential ROS-mediated adverse effects26-29 including increased susceptibility to infections30, 31 and renal toxicity in patients32, 33 and in rats34 but not reported in sponsored rat studies using a similar dose.

There is little doubt that iron causes oxidative stress and the clinical literature is substantial, differing reports and opinions regarding its clinical significance still being commonplace35-40. However, several investigators have pointed out that the clinical impact of experimental findings is largely hypothetical and speculative since patient safety data for NPI products are generally reassuring provided that the recommended dosing regimen is not exceeded and overdose avoided41-43. NPI products that contain free or labile iron, or release iron by rapid degradation of the carbohydrate coating, are considered more likely to produce adverse effects (by transferrin supersaturation and/or ROS generation)44,45; the maximum safe clinical doses of different NPI products seem to be governed by this phenomenon. By contrast, a recent report46 suggests that “naked” iron in the form of IV ferric chloride has a similar clinical safety profile to iron sucrose but is not quite as effective. Overall, ROS generation appears to be not associated with adverse outcomes47, 48, although caution in some situations, such as in patients with sepsis and chronic kidney disease patients receiving haemodialysis, may be appropriate.

The toxicological evaluation recommended by SWP for all NPI products, irrespective of whether appropriate physicochemical standards are met, is essentially a comparative iron-distribution study with timed sampling of plasma, the reticuloendothelial system and target tissues for iron content. No results from such a testing paradigm were cited in the reflection paper and no data could be found in the literature on generic iron sucrose preparations. Thus, it appears that the recommended approach is entirely hypothesis-based, unvalidated and with no established correlation to clinical safety. Moreover, critical information is lacking on dosing regimen, interpretation criteria and inherent variability (in respect of distinguishing between different batches of originator iron sucrose and USP-compliant and non-USP-compliant iron sucrose). On the other hand, a generic iron sucrose product, Iron Sucrose Azad, that meets the USP specification in all respects, is reported to have a closely similar toxicological profile to that of originator iron sucrose49.

The proposed testing regimen is somewhat analogous to the comparative toxicity assessment of biosimilar products50, but the latter are much more difficult, if not impossible, to characterise using physicochemical data alone and there are sufficient checks and balances in the overall safety programme to evaluate the clinical relevance of any apparent toxicological differences. A generic iron sucrose is included as a candidate for in vivo assessment in a publication on the therapeutic equivalence of complex drugs51. Because only sponsored publications are used as support for this approach and since one of the co-authors is an employee of the originator company, the justification presented on generic iron sucrose is considered to be highly compromised.

The reflection paper fails to include two significant regulatory precedents. A generic iron sucrose product, Ferrologic, was awarded EU approval in 2007/8 based solely on comparative physiochemical data/USP requirements52 and an entirely new NPI product, Monofer, was approved in 2009 in the absence of drug-specific toxicological data (using “read-across” from data on iron dextran)53. Thus, the SWP is requesting a significant toxicological evaluation of generic NPIs whereas two precedents suggest comparative physicochemical data or read-across have in the past been considered sufficient. This insistence on a toxicological evaluation is contrary to the spirit of the existing animal welfare provisions of EU legislation54, which are soon to be strengthened by Directive 2010/63/EU on the protection of animals used for scientific purposes. Article 4.1 of this directive indicates that member states should ensure that, wherever possible, a scientifically satisfactory method or testing strategy, not entailing the use of live animals, shall be used instead of a procedure55.

Regulatory omissions

The content, recommendations and the status of the reflection paper raise a number of critically important regulatory questions.

Firstly, when preparing “guidance” documents, should the SWP be entitled to ignore requirements applied to applicants in Directive 2001/83/EC56 on medicines for human use in terms of including all relevant information, whether favourable or unfavourable (Annex 1, General Principles), and preparing an overview that contains an integrated and critical assessment of the non-clinical data and a discussion and justification of the testing strategy (Annex 1, 2001/83, PI, 2.4)?

Why did the SWP fail to mention the provisions of Annex 1, PI, section 3.2 (6) indicating that if no relevant monograph is available in the European Pharmacopoeia nor in the pharmacopoeia of a member state, “compliance with the monograph of a third country pharmacopoeia can be accepted”?

Why, when such a reflection paper amounts to de facto regulatory guidance, was there no inbuilt public consultation? (It would be somewhat disingenuous for the SWP to argue that individual applicants can challenge the provisions of the reflection paper since only the bravest and most confident assessor would be willing to contradict an opinion from the working party.)

Is it justified to dismiss largely by assertion rather than detailed scientific justification an approach based on physicochemical data, the latter being supported by published data and precedents, thereby apparently ignoring existing and additional impending requirements on minimisation of procedures employing experimental animals?

Is the recommendation to employ a completely untested and unvalidated toxicological paradigm with no proven correlation to clinical safety justified?

If no significant differences were detected between the originator NPI product and a generic NPI using the recommended testing paradigm, would this overrule any potential quality deficiencies related to drug substance specification criteria? (If not, this is considered to further undermine the rationale for in vivo testing.)

Finally, is it acceptable to require toxicological studies on a generic version of an originator product that gained approval without any compound-specific non-clinical evaluation? The latter situation, a complete inversion of the normal legal requirements on generic products, could arise in the future in relation to generic versions of Monofer.

In view of these numerous concerns, the SWP may wish to reconsider its position on the reflection paper. Otherwise, it could be inferred that the working party is not averse to setting a general regulatory precedent in relation to the omission of key evidence on drug quality and safety and to the acceptability of assessments that are incomplete and potentially misleading.

References

1. EMA, Reflection paper on non-clinical studies for generic nanoparticle iron medicinal product applications, EMA/CHMP/SWP/100094/2011, 17 March 2011, www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2011/04/WC500105048.pdf

2. Wang YX, Hussain SM, Krestin GP, Superparamagnetic iron oxide contrast agents: physicochemical characteristics and applications in MR imaging, Eur Radiol, 2001, 11(11):2319-31

3. Laurent S, Boutry S, Mahieu I, Vander Elst L, Muller RN, Iron oxide based MR contrast agents: from chemistry to cell labelling, Curr Med Chem, 2009, 16(35):4712-27

4. Pouliquen D, Le Jeune JJ, Perdrisot R, Ermias A, Jallet P, Iron oxide nanoparticles for use as an MRI contrast agent: pharmacokinetics and metabolism, Magn Reson Imaging, 1991, 9(3):275-83

5. Weissleder R, Stark DD, Engelstad BL, Bacon BR, Compton CC, White DL, Jacobs P, Lewis J, Superparamagnetic iron oxide: pharmacokinetics and toxicity, AJR Am J Roentgenol, January 1989, 152(1):167-73

6. Auerbach M, Ballard H. Clinical use of intravenous iron: administration, efficacy, and safety, Hematology Am Soc Hematol Educ Program, 2010, 2010:338-47

7. Ibid

8. Venofer (iron sucrose), Summary of Product Characteristics: http://www.medicines.org.uk/emc/medicine/24168/SPC

9. Venofer (iron sucrose injection, USP), FDA label: www.accessdata.fda.gov/drugsatfda_docs/label/2007/021135s017lbl.pdf

10. Iron Sucrose Injection, USP 29 Monograph: www.pharmacopeia.cn/v29240/usp29nf24s0_m42475.html

11. See reference 6

12. Van Wyck D, Anderson J, Johnson K, Labile iron in parenteral iron formulations: a quantitative and comparative study, Nephrol Dial Transplant, March 2004, 19(3):561-5

13. Danielson BG. Structure, chemistry, and pharmacokinetics of intravenous iron agents, J Am Soc Nephrol, December 2004, 15 Suppl 2:S93-8

14. Roth S, Langguth P, Spicher K, Enzmann H, Comparative toxicity and cell-tissue distribution study on nanoparticular iron complexes using avian embryos and HepG2-cells, Transl Res, January 2008, 151(1):36-44

15. Toblli JE, Cao G, Oliveri L, Angerosa M, Differences between original intravenous iron sucrose and iron sucrose similar preparations, Arzneimittelforschung, 2009, 59(4):176-90

16. Karlsson HL, Gustafsson J, Cronholm P, Möller L, Size-dependent toxicity of metal oxide particles – a comparison between nano- and micrometer size, Toxicol Lett, 24 July 2009, 188(2):112-8

17. Toblli JE, Cao G, Oliveri L, Angerosa M. Differences between original intravenous iron sucrose and iron sucrose similar preparations, Arzneimittelforschung, 2009, 59(4):176-90

18. Karlsson HL, Gustafsson J, Cronholm P, Möller L. Size-dependent toxicity of metal oxide particles – a comparison between nano- and micrometer size, Toxicol Lett, 24 July 2009, 188(2):112-8

19. Toblli JE, Cao G, Oliveri L, Angerosa M, Differences between the original iron sucrose complex Venofer and the iron sucrose similar Generis, and potential implications, Port J Nephrol Hypert, 2009, 23(1): 53-63

20. Bailie, GR, Iron sucrose and iron sucrose similar (ISS): what are the significant differences?, www.aapspharmaceutica.com/meetings/files/172/Session_IIc_-_Bailie_(yes-yes).pdf

21. Vu'o'ng Lê B, Khorsi-Cauet H, Villegier AS, Bach V, Gay-Quéheillard J, New rat models of iron sucrose-induced iron overload, Exp Biol Med (Maywood), 1 July 2011, 236(7):790-9

22. Rottembourg J, Kadri A, Leonard E, Dansaert A, Lafuma A, Do two intravenous iron sucrose preparations have the same efficacy?, Nephrol Dial Transplant, 25 February 2011 [E pub]

23. Fer Mylan (exceptional approval), http://afssaps-prd.afssaps.fr/php/ecodex/notice/N0185893.htm

24. Regulation of Complex Molecules: Vifor Roundtable Report, www.hospitalpharmacyeurope.com/editorial/attachment.asp?aaid=892

25. Sturm B, Laggner H, Ternes N, Goldenberg H, Scheiber-Mojdehkar B, Intravenous iron preparations and ascorbic acid: effects on chelatable and bioavailable iron, Kidney Int, March 2005, 67(3):1161-70

26. Pai AB, Boyd AV, McQuade CR, Harford A, Norenberg JP, Zager PG, Comparison of oxidative stress markers after intravenous administration of iron dextran, sodium ferric gluconate, and iron sucrose in patients undergoing hemodialysis, Pharmacotherapy, March 2007, 27(3):343-50

27. Pai AB, Conner T, McQuade CR, Olp J, Hicks P, Non-transferrin bound iron, cytokine activation and intracellular reactive oxygen species generation in hemodialysis patients receiving intravenous iron dextran or iron sucrose, Biometals, August 2011, 24(4):603-13

28. Zager RA, Johnson AC, Hanson SY, Wasse H, Parenteral iron formulations: a comparative toxicologic analysis and mechanisms of cell injury, Am J Kidney Dis, July 2002, 40(1):90-103

29. Zager RA, Parenteral iron compounds: potent oxidants but mainstays of anemia management in chronic renal disease, Clin J Am Soc Nephrol, September 2006, 1 Suppl 1:S24-31

30. Gupta A, Zhuo J, Zha J, Reddy S, Olp J, Pai A, Effect of different intravenous iron preparations on lymphocyte intracellular reactive oxygen species generation and subpopulation survival, BMC Nephrol, 17August 2010, 11:16

31. Barton Pai A, Pai MP, Depczynski J, McQuade CR, Mercier RC, Non-transferrin-bound iron is associated with enhanced Staphylococcus aureus growth in hemodialysis patients receiving intravenous iron sucrose, Am J Nephrol, 2006, 26(3):304-9

32. Bishu K, Agarwal R, Acute injury with intravenous iron and concerns regarding long-term safety, Clin J Am Soc Nephrol, September 2006, 1 Suppl 1:S19-23

33. Agarwal R, Leehey DJ, Olsen SM, Dahl NV, Proteinuria induced by parenteral iron in chronic kidney disease – a comparative randomized controlled trial, Clin J Am Soc Nephrol, January 2011, 6(1):114-21

34. Breborowicz A, Polubinska A, G�rna K, Breborowicz M, Oreopoulos DG, Iron sucrose induced morphological and functional changes in the rat kidney, Transl Res, November 2006, 148(5):257-62

35. Van Buren P, Velez RL, Vaziri ND, Zhou XJ, Iron overdose: a contributor to adverse outcomes in randomized trials of anemia correction in CKD, Int Urol Nephrol, 10 July 2011

36. See reference 27

37. Stef�nsson BV, Haraldsson B, Nilsson U, Acute Oxidative Stress following Intravenous Iron Injection in Patients on Chronic Hemodialysis: A Comparison of Iron-Sucrose and Iron-Dextran, Nephron Clin Pract, 2011, 118(3):c249-56

38. Rangel EB, Esp�sito BP, Carneiro FD, Mallet AC, Matos AC, Andreoli MC, Guimarães-Souza NK, Santos BF, Labile plasma iron generation after intravenous iron is time-dependent and transitory in patients undergoing chronic hemodialysis, Ther Apher Dial, April 2010, 14(2):186-92

39. Garneata L, Intravenous iron, inflammation, and oxidative stress: is iron a friend or an enemy of uremic patients?, J Ren Nutr, January 2008, 18(1):40-5

40. Agarwal R, Iron, oxidative stress, and clinical outcomes, Pediatr Nephrol, August 2008, 23(8):1195-9.

41. Hörl WH, Iron therapy for renal anemia: how much needed, how much harmful?, Pediatr Nephrol, April 2007, 22(4):480-9

42. Malindretos P, Sarafidis PA, Rudenco I, Raptis V, Makedou K, Makedou A, Grekas DM, Slow intravenous iron administration does not aggravate oxidative stress and inflammatory biomarkers during hemodialysis: a comparative study between iron sucrose and iron dextran, Am J Nephrol, 2007, 27(6):572-9

43. Scheiber-Mojdehkar B, Lutzky B, Schaufler R, Sturm B, Goldenberg H, Non-transferrin-bound iron in the serum of hemodialysis patients who receive ferric saccharate: no correlation to peroxide generation, J Am Soc Nephrol, June 2004, 15(6):1648-55

44. Beshara S, Lundqvist H, Sundin J, Lubberink M, Tolmachev V, Valind S, Antoni G, Långström B, Danielson BG, Pharmacokinetics and red cell utilization of iron(III) hydroxide-sucrose complex in anaemic patients: a study using positron emission tomography, Br J Haematol, February 1999, 104(2):296-302

45. Geisser P, Burckhardt S, The Pharmacokinetics and Pharmacodynamics of Iron Preparations, Pharmaceutics, 2011, 3, 12-23

46. Wu CJ, Lin HC, Lee KF, Chuang CK, Chen YC, Chen HH. Comparison of parenteral iron sucrose and ferric chloride during erythropoietin therapy of haemodialysis patients, Nephrology (Carlton), February 2010, 15(1):42-7

47. See reference 8

48. See reference 9

49. Meier T, Schropp P, Pater C, Leoni AL, Khov-Tran VV, Elford P, Physicochemical and toxicological characterization of a new generic iron sucrose preparation, Arzneimittelforschung, 2011, 61(2):112-9

50. Guideline on similar biological medicinal products containing biotechnology-derived proteins as active substance: non-clinical and clinical issues, EMEA/CHMP/BMWP/42832/2005, www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2009/09/WC500003920.pdf

51. Schellekens H, Klinger E, Mühlebach S, Brin JF, Storm G, Crommelin DJ, The therapeutic equivalence of complex drugs, Regul Toxicol Pharmacol, February 2011, 59(1):176-83

52. Public Assessment Report for Ferrologic, www.lakemedelsverket.se/SPC_PIL/Pdf/par/Ferrologic%20solu%20f%20inj%20conc%20f%20solu%20f%20inf.pdf

53. Public Assessment Report for Monofer, www.lakemedelsverket.se/SPC_PIL/Pdf/par/Monofer%20solution%20for%20infusion-injection.pdf

54. Directive 86/609/EEC, http://eur-lex.europa.eu/LexUriServ/LexUriServ.do?uri=CELEX:31986L0609:EN:HTML

55. Concept paper on the Need for Revision of the Position on the Replacement of Animal Studies by in vitro Models (CPMP/SWP/728/95), www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2011/04/WC500105110.pdf

56. Directive 2001/83/EC, http://ec.europa.eu/health/files/eudralex/vol-1/dir_2001_83_cons2009/2001_83_cons2009_en.pdf

Dr David Snodin is the principal of Xiphora Biopharma Consulting, a UK-based consultancy offering nonclinical services to the pharmaceutical sector. Dr Snodin is also a former expert preclinical assessor at the UK Medicines Control Agency (now the Medicines and Healthcare products Regulatory Agency) and a former UK representative on the EMA/CHMP safety working party. Website: http://xiphora.com/. Email: [email protected].

Latest Headlines
See All
UsernamePublicRestriction

Register

PS116347

Ask The Analyst

Ask the Analyst is free for subscribers.  Submit your question and one of our analysts will be in touch.

Your question has been successfully sent to the email address below and we will get back as soon as possible. my@email.address.

All fields are required.

Please make sure all fields are completed.

Please make sure you have filled out all fields

Please make sure you have filled out all fields

Please enter a valid e-mail address

Please enter a valid Phone Number

Ask your question to our analysts

Cancel