Skip to main content

Biosimilar and Biobetter Scenarios for the US and Europe: What Should We Expect?

  • Chapter
Biobetters

Part of the book series: AAPS Advances in the Pharmaceutical Sciences Series ((AAPS,volume 19))

Abstract

We explore differences between biosimilars and conventional small molecule pharmaceutical generic drugs with respect to development, regulatory requirements, legal challenges, pricing and related market dynamics over the product life cycle, and compare these dynamics in the U.S. and Europe. We also address issues concerning the development, manufacturing and marketing of biobetters as well as biosimilars. After identifying a number of measurement issues involving quantification of medicine utilization volumes, using IMS MIDAS™ data we quantify adoption of biosimilars in nine European countries between 2006 and 2012. We discuss factors affecting investment decisions among traditional novel pharmaceuticals, novel biologics, biosimilars and biobetters, and describe various strategies recently employed by biopharmaceutical firms. We conclude by considering issues involving immunogenicity of biologics, as well as controversies regarding whether biosimilars should have identical international non-proprietary names as the reference brand.

Ernst R. Berndt is the Louis E. Seley Professor in Applied Economics, and Mark R. Trusheim is Visiting Scientist, both at the Massachusetts Institute of Technology, Alfred P. Sloan School of Management, Cambridge, Massachusetts, USA.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 129.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 169.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 169.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Notes

  1. 1.

    Trusheim et al. (2010).

  2. 2.

    Grabowski et al. (2011) define a complex small molecule as one meeting two or more of the following criteria: black box warning, narrow therapeutic index, prescribed by specialists, oncology products, or manufacturing technology that is available to only a limited number of firms. The more complex the drug, ceteris paribus, the fewer the number of generic entrants at the time of LOE. Olson and Wendling (2013) find that entry both during and after the 180-day exclusivity depends not only on pre-LOE market size, but also is greater if the drug at issue was originally designated a New Chemical Entity (NCE) by the Food and Drug Administration during the NDA approval process.

  3. 3.

    IMS Institute for Healthcare Informatics (2011a), p. 21, and Aitken et al. (2013). Earlier studies include those by Hurwitz and Caves (1988), Ellison et al. (1997), Cook (1998), Reiffen and Ward (2005), Saha et al. (2006), Aitken et al. (2008), Aitken and Berndt (2011), Berndt and Aitken (2011) and Berndt and Newhouse (2012). Regarding complexity, as discussed in the previous endnote, Grabowski et al. (2011, pp. 540–541) report that “On average, drugs with two or more characteristics faced 2.5 geneic entrants 1 year following initial generic entry, while drugs with one or no complexity characteristics faced an average of 8.5 entrants.” Mean generic share of non-complex small molecules was 1.7 times larger than for complex small molecules, while the mean price discount from brand price was 1.6 times larger (price here reflecting manufacturer’s revenues from sales to wholesalers and direct customers).

  4. 4.

    If multiple Paragraph IV filers submit their challenge on the same day, the various challengers share the 180-day exclusivity, resulting in a larger number of competitors. See Federal Trade Commission (2011, especially Chap. 7) for more details.

  5. 5.

    See Federal Trade Commission (2002, 2011), and Aitken et al. (2013) for further details.

  6. 6.

    Berndt et al. (2007). Also see Branstetter et al. (2011) for estimated effects of Paragraph IV entry on consumers’ welfare in the U.S.

  7. 7.

    Aitken et al. (2013).

  8. 8.

    Berndt et al. (2007); Federal Trade Commission (2011); Aitken et al. (2013).

  9. 9.

    Federal Trade Commission (2011, Chaps. 2 and 7, Aitken et al. (2013), Grabowski et al. (2014), and Drake et al. (2014).

  10. 10.

    See Aitken et al. (2008) for details.

  11. 11.

    Berndt and Aitken (2011); Reiffen and Ward (2007).

  12. 12.

    IMS Institute for Healthcare Informatics (2011a, p. 21).

  13. 13.

    IMS Institute for Healthcare Informatics (2014, p. 40).

  14. 14.

    For a discussion of the lucrative profitability of a successful Paragraph IV first to file challenger, even if the brand responds by launching an authorized generic, see, for example, Drug Channels (2011, 2012).

  15. 15.

    Berndt and DuBois (2012) amend the Danzon and Furukawa (2011) classification slightly, characterizing France as becoming pharmacy driven in 2006 (when pharmacies were first given strong financial incentives to substitute generics for brands) and Germany in 2007.

  16. 16.

    According to the FDA (2014b, p. 15), biosimilarity means that “the biological product is highly similar to the reference product notwithstanding minor differences in clinically inactive components,” and that “there are no clinically meaningful differences between the biological product and the reference product in terms of the safety, purity and potency of the product.” Regarding interchangeability decisions, in the same FDA (2014b, pp. 5–6) document, draft guidance specified four possible process decision outcomes: definitely not interchangeable; might be interchangeable if enough clinical and analytical data to support it is developed; looks like it is interchangeable but needs more clinical and analytical data to support it; and biosimilar is a “fingerprint” of the innovator product, as demonstrated by analytical data, and no further clinical data is required.

  17. 17.

    Trusheim et al. (2010).

  18. 18.

    Leeson (2012).

  19. 19.

    Schellekens (2005).

  20. 20.

    Ziegler and Santagostino (2011).

  21. 21.

    Kelley (2009).

  22. 22.

    Morrow (2006).

  23. 23.

    European Medicines Agency (2005).

  24. 24.

    The product-specific biosimilar guidelines include recombinant erythropoietins, low-molecular-weight heparins, recombinant interferon alpha, recombinant granulocyte-colony stimulating factor, somatropin, recombinant human insulin, and monoclonal antibodies. See European Medicines Agency (2006a, b, c, 2009a, b, 2010, 2012a, b, c).

  25. 25.

    European Medicines Agency (2010, 2011a, b, c, 2012a, c (a finalized version of 2010))

  26. 26.

    European Medicines Agency (2013a).

  27. 27.

    European Medicines Agency (2013a, p. 4). BioPartners’ Valtropin was approved by the EU on the same day as Sandoz’ Omnitrope with Humatrope as the reference product, but Valtropin has not been marketed.

  28. 28.

    European Medicines Agency (2012b).

  29. 29.

    European Medicines Agency (2012b).

  30. 30.

    Ehmann (2010).

  31. 31.

    European Commission (2013, p. 27).

  32. 32.

    Biotechnology Industry Association (2013). For additional discussion, see Karst (2013a), who also provides a state-by-state legislation scorecard.

  33. 33.

    Grabowski et al. (2013, p. 3).

  34. 34.

    Aitken et al. (2008), Berndt and Aitken (2011) and Aitken et al. (2013).

  35. 35.

    Conti and Berndt (2014).

  36. 36.

    World Health Organization (2009, pp. 1–2); italics and bold in original text. For further details concerning DDD, see World Health Organization (2003 (Chap. 6), 2011); also see International Federation of Pharmaceutical Manufacturers and Associations (2006).

  37. 37.

    From email correspondence with Terry McMonagle at the IMS Institute for Healthcare Informatics, September 4, 2013, 11:15 am.

  38. 38.

    See footnote 37.

  39. 39.

    See footnote 37.

  40. 40.

    See Walsh (2013) for examples and discussion.

  41. 41.

    The analyst’s comments are referenced in the note below Table 2 of this document; also see Walsh (2013).

  42. 42.

    We note in passing that the 81 % biosimilar share reported for filgrastim in the UK in 2012 in Table 3 based on standard unit measures is very similar to the part-year 2013 83 % share for filgrastim in the UK reported in Walsh (2013, slide 8).

  43. 43.

    The dollar and standard unit shares do not show complete concordance for Finland and Norway. The underlying IMS MIDAS data shows some minimal reference product dollar sales in spite of zero standard unit shipments. This could be due to reporting timing differences or other idiosyncratic causes affecting the very small values.

  44. 44.

    We note, however, that in our nine country sample, for somatropin and filgrastim, only in Spain is dispensing limited to the hospital setting; in all other countries for these two molecules, dispensing occurs in both the hospital and retail setting. For erythropoietin alpha, in both Spain and Belgium dispensing occurs only in the hospital setting, and in all other countries dispensing takes place in both the hospital and retail settings.

  45. 45.

    Aitken et al. (2013), and Berndt and Dubois (2012).

  46. 46.

    European Commission (2013, pp. 32–33).

  47. 47.

    As quoted in FiercePharma (2014).

  48. 48.

    Staton (2014).

  49. 49.

    Staton (2014).

  50. 50.

    Karst (2013b),

  51. 51.

    As quoted in Berkrot (2013).

  52. 52.

    See footnote 51.

  53. 53.

    See footnote 51.

  54. 54.

    Carroll (2013a, b).

  55. 55.

    As quoted in Carroll (2011).

  56. 56.

    Carroll (2013c).

  57. 57.

    McBride (2012).

  58. 58.

    McBride (2013).

  59. 59.

    Palmer (2013).

  60. 60.

    As quoted in pmlive.com (2013).

  61. 61.

    Gardner (2013).

  62. 62.

    DiMaisi et al. (2003).

  63. 63.

    DiMasi and Grabowski (2007).

  64. 64.

    Trusheim et al. (2011).

  65. 65.

    Trusheim and Berndt (2012).

  66. 66.

    Baird et al. (2013).

  67. 67.

    According to Brennan (2014), as of June 2014 24 countries have established biosimilar pathways or have approved follow-on biologics.

  68. 68.

    Epoietinalfa, June 24, 2011 Division Director Summary Review, STN BL 103234/5166, p. 5 of 38. Available online at Drugs@FDA.

  69. 69.

    Grabowski et al. (2013, p. 4).

  70. 70.

    IMS Health (2011), slides 10, 11 and 40.

  71. 71.

    Drug Facts and Comparisons (2011), p. 154.

  72. 72.

    Grabowski (2013, slide 5).

  73. 73.

    Genotropin, FDA Center for Drug Evaluation and Research, Application Number 20-280/S-031, Review—Administrative Documents, dated July 23, 2001. Available online at Drugs@FDA, Approval History, NDA 020280, 07/25/2001 031, p. 1 of 3, letter from David G. Orloff, M.D., Director, Division of Metabolic and Endocrine Drug Products to file NDA 20-280/S-031.

  74. 74.

    “Somatropin”, Drug Facts and Comparisons (2011), pp. 523–526.

  75. 75.

    Genotropin Draft Package Insert, p. 3of 15, NDA 20-280/S-031, available online at Drugs@FDA, Approval History, NDA 020280, 07/25/2001.

  76. 76.

    IMS Health (2011), slide 41.

  77. 77.

    Neupogen, “Review and Summary Basis of Approval”, Application Number 103353/000, Center for Drug Evaluation and Research, February 20, 1991. “Supplement 1036, Letter, April 2, 1998. Available online at Drugs@FDA.

  78. 78.

    “Filgrastim”, Drug Facts and Comparisons (2011), p. 163.

  79. 79.

    “Neutropenia”, in White Blood Cell Disorders, ch. 158, in Robert Berkow, Editor, The Merck Manual of Medical Information, (Home Edition), Whitehouse Station, NJ: Merck Research Laboratories, 1998, pp. 761–763.

  80. 80.

    “Neupogen (Filgrastim) Drug Information: User Reviews, Drug Side Effects…”, p. 1 of 3 in RxList…, last reviewed June 4, 2012. b Available online at http://www.rxlist.com/neupogen-drug.htm.

  81. 81.

    Kim et al. (2003), p. 1 of 14.

  82. 82.

    IMS Health (2011), slide 42. According to Grabowski et al. (2013, p. 4), Ratiopharm’s Filgrastim ratiopharm was approved by the EU on September 15, 2008, but was withdrawn on July 20, 2011.

  83. 83.

    “Pegfilgrastim”, Drug Facts and Comparisons (2011), p. 166.

  84. 84.

    “Ankylosing spondylitis” in Anderson et al. (1998), pp. 94–95.

  85. 85.

    Drug Facts and Figures (2011), pp. 2822–2823.

  86. 86.

    “Folliropin Alpha” Drug Facts and Comparisons (2009), p. 350.

  87. 87.

    European Medicines Agency (2013c).

  88. 88.

    Food and Drug Administration (2014a).

References

Download references

Acknowledgements

The research reported on here was funded by an educational grant from Pfizer Limited, Surrey, UK to Berndt Associates LLC. The funding source had no role in the design and conduct of the study; collection, management, analysis, or interpretation of the data; and while it provided comments on a draft version of this manuscript, it had no role in the preparation, review, submission, or approval of the manuscript for publication. The authors thank Kirsten Axelsen, Adam Heathfield, Jake Lebiecki and Danielle Rollman of Pfizer for comments on an earlier draft of this manuscript, and participants at the TIGER Forum 2014 at the Toulouse School of Economics in Toulouse, France, June 2, 2014. The statements, findings, conclusions, views, and opinions contained and expressed herein are those of the authors and are based in part on IMS MIDAS™ data obtained by Berndt Associates LLC under license from IMS Health (rights reserved), and are not necessarily those of IMS Health, its affiliates or subsidiaries, or the institutions with whom the authors are affiliated. Any errors or misstatements are our own.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Ernst R. Berndt .

Editor information

Editors and Affiliations

Appendix

Appendix

Biologic Molecules with Biosimilar Entry in Europe and Elsewhere

Short-Acting Epoietin Recombinant (Erythropoietin, Alpha, Beta, Theta, Zeta)

According to the US Food and Drug Administration, “Erythropoietin is a glycoprotein whose main function is to stimulate the proliferation and differentiation of erythroid precursors in the bone marrow. Erythropoietin is produced mainly in the kidneys, though several other tissues produce lesser amounts of the growth factor.”Footnote 68 Approved by the FDA on June 1, 1989, Epogen/Procrit (epoetinalfa) was produced in Chinese Hamster Ovary cells that have modified through recombinant DNA technology to encode the gene for human erythropoietin, and was initially approved for the treatment of anemia in patients with chronic renal failure. Epogen/Procrit was subsequently approved for the treatment of anemia due to ziduvodine therapy in HIV-infected patients (1991) and for the treatment of anemia in patients with non-myeloid malignancies whose anemia is due to the effect of concomitantly administered chemotherapy (1993). Both these supplemental approvals were based on demonstration of a reduction in the proportion of patients receiving shaded blood cell (RBC) transfusions.

Within Europe, the epoietin alpha reference product is Johnson & Johnson’s Erypo or Eprex. As of June 2011, there were five approved biosimilar products: Binocrit (Sandoz/Novartis), Epo A (Hexal/Novartis), Abseamed (Medici), Retacrit (Hospira) and Silapo (Stada), all approved between August 28, 2007 and December 18, 2007.Footnote 69 Using the IMS Health classification scheme, there are two non-referenced products in the epoietin biosimilar accessible market (defined as an original product, granted market exclusivity at the start of its commercial life in Europe, whose exclusivity is now expired, with the product never have been referenced, or may have been referenced but the referencing biosimilar has not yet launched): Roche’s NeoRecormon, and Teva’s Eporatio/Biopoin.Footnote 70 In the U.S., in addition to having been approved by the FDA for treating anemia in cancer patients on chemotherapy, anemia in chronic renal failure patients, and anemia in zidovudine-treated, HIV-infected patients, epoietin alfa is approved for the reduction of allogenic blood transfusion in surgery patients. As of 2011, Epogen/Procrit was available in 2000, 3000 and 4000 units/ml 1 ml single-dose vials for subcutaneous injection or intravenous solution administration, in 20000 units/ml 1 ml multidose vials, in 10000 units/ml 1 ml single-dose and 2 ml multidose vials, and in 40000 units/ml single-dose vials for subcutaneous injection or intravenous solution administration.

Another erythropoietin stimulating agent (ESA) approved in both the US and EU is Amgen’s darbepoetin alfa (brand name Aranesp in the US). Darbepoietin is distinguished from epoietin agents primarily because of Aranesp’s longer serum half-life, implying generally less frequent dosing than the epoietins.Footnote 71 Currently Aranesp is patent-protected in the US and EU, with its earliest reported year of key US patent expiry being 2024; for Epogen this US patent expiry date is 2013.Footnote 72

Growth Hormone for Children Born Small for Gestational Age—SGA (Somatropin Molecule)

Of the approximately 2.5 % of children who are born small for gestational age (SGA), 10–15 % fail to “catch up” by age two. Children who do not catch up by age two, if left untreated, are destined in many cases to have compromised final height, relative to the norm for the population. A relative height measure is SDS—the number of standard deviations an individual at a particular age is away from the age-specific population mean. Though there are differences across and within countries on the measure of SDS triggering treatment, growth hormone supplementation in children born SGA can enhance growth velocity, height SDS, and predicted adult height. Aside from the known adverse effects of growth hormone therapy, of concern in treating these children is a risk of accelerating bone age beyond chronological age, with the possibility of precipitating precocious puberty and compromising final stature on that basis.Footnote 73

On August 24, 1995, the FDA approved Pharmacia and Upjohn’s NDA # 020280 application for Genotropin (somatropin recombinant) for children born SGA who fail to manifest catch-up growth by 2 years of age, caused by an inadequate secretion of endogenous growth hormone. Over the years the FDA has approved a number of supplemental indications (e.g., growth failure associated with chronic renal insufficiency, with Noonan syndrome, with Prader-Willi syndrome, with Turner syndrome, in adults with either adult- or childhood-onset growth hormone deficiency, and others) as well as several related somatropin products, such as Omnitrope (Sandoz), Serostim (Serono), Humatrope (Eli Lilly), Nutropin (Genentech), Salzen (Serono), Tev-Tropin (Gate), HumatroPen (Eli Lilly), Zorbtive (Serono), Norditropin (Novo Nordisk), Accretropin (Cangene), and Nutropin AQ, Nutropin AQ NuSpin 5, NuSpin 10 and NuSpin 20 (Genentech). Most of these formulations are subcutaneous injection, lyophilized power for solution, although some products, such as Norditropin, involve pen or two-chamber cartridge delivery systems, with a reconstitution device used to mix the diluent and powder.Footnote 74 Somatropin must not be injected intravenously. Genotropin lyophilized powder contains somatropin of rDNA origin, a polypeptide hormone. The amino acid sequence of the product is identical to that of human growth hormone of pituitary origin (somatropin). Genotropin is synthesized in a strain of Escherichia coli that has been modified by the addition of the gene for human growth hormone.Footnote 75

According to IMS Health, as of June 2011 both Pfizer’s (who acquired rights with the acquisition of Pharmacia and Upjohn) Genotropin and Eli Lilly’s Humatrope were reference products, the two approved biosimilar products were Novartis Sandoz’ Omnitrope and Somatropin (unknown manufacturing laboratory), whereas the non-referenced products included Sanofi Aventis’ Maxomat, Nova Nordisk’s Norditropin, Ipsen’s Nutropinaq, Merck Serono’s Saizen, and Ferring’s Zomacton.Footnote 76

Granulocyte-Colony Stimulating Factor (G-CSF), Filgrastim and Lenograstim Molecules

On February 20, 1991, the US FDA approved Amgen’s filgrastim (trade name Neupogen) to decrease the incidence of infection, as manifested in febrile neutropenia, in patients with non-myeloid malignancies receiving myelosuppressive anti-cancer drugs associated with a significant incidence of severe neutropenia with fever. On April 2, 1998, the FDA granted a supplemental NDA approval for acute myeloid leukemia (AML) adult patients receiving induction or consolidation chemotherapy, for reducing the time to neutrophil recovery and the duration of fever.Footnote 77 Other approvedFootnote 78 uses of filgrastim include in patients with nonmyeloid malignancies undergoing myeloablative chemotherapy followed by bone marrow transplantation, and for the mobilization of hematopoietic progenitor cells into the peripheral blood for collection by leukapheresis.

Neutropenia is condition with an abnormally low number of neutrophils in the blood—the body’s primary cellular defense system against bacteria and fungi. Neutrophils also help heal wounds and ingest foreign debris, such as embedded splinters. People who have severe neutropenia (fewer than 500 neutrophils per microliter of blood) can rapidly succumb to infection because their bodies lack the means to fight the invading organisms. Neutrophils mature in the bone marrow in about 2 weeks. After entering the blood stream, they circulate for about 6 h, searching for infective organisms and other intruders. When they find one, they migrate into the tissues, attach themselves to the intruders, and produce toxic substances that kill and digest the intruders. This reaction may damage healthy tissue in the area of the infection. The entire process produces an inflammatory response in the infected area, which appears on the body’s surface as redness, swelling, and heat. Neutropenia has several causes. The number of neutrophils can decrease because bone marrow production isn’t adequate or because large numbers of white blood cells are destroyed in the circulation. Aplastic anemia, and certain rare genetic diseases such as infantile genetic agranulocytosis and familial neutropenia cause decreases in the number of white blood cells. Certain drugs, especially chemotherapies used in cancer treatment, impair the bone marrow’s ability to produce neutrophils. Growth factors that stimulate the production of white blood cells, particularly granulocyte colony-stimulating factor (G-CSF) and granulocyte-macrophage colony-stimulating factor (GM-CSF) can eliminate neutropenic episodes in cyclic neutropenia.Footnote 79

In the U.S., Neupogen is still marketed exclusively by Amgen. Four dosage forms are approved—two in single use vials, and two as pre-filled injectable syringes. Neupogen is produced by Escherichia coli (E coli) bacteria into which has been inserted the human granulocyte colony-stimulating factor gene. The protein has an amino acid sequence that is identical to the natural sequence predicted from human DNA analysis, except for the addition of an N-terminal methionine necessary for expression in E coli. Because Neupogen is produced in E coli, the product is non-glycosylated and thus differs from G-CSF isolated from a human cell. Neupogen is a sterile, clear, colorless preservative-free liquid for parenteral administration. In order to maintain clinical benefit, chronic daily administration is required.Footnote 80

A second form of recombinant human granulocyte colony-stimulating factor is lenograstim (brand name Granocyte, Chugai Pharmaceuticals, marketed in the EU by Sanofi Aventis), a Chinese hamster ovary-derived G-CSF, indistinguishable from native G-CSF, and differing from filgrastim which is an Escherichica coli-derived G-CSF and is non-glycosylated, having an extra methionine group at the N-terminal end of the peptide chain.Footnote 81 Granocyte (lenograstim) is not available in the U.S., but is a non-referenced product in the EU, having brand names in addition to Sanofi Aventis’ Granocyte, Euprotin (Almirall), Myelostim (Italfarmaco) and Roche’s Neutrogin. As of June 2011, a number of biosimilars were approved in the EU using Amgen’s filgrastim (Neupogen) as the reference product. These biosimilars include a Biograstim (CT Arzzneimittel), Novartis’/Sandoz filgrastim Zarzio, Teva’s Tevagrastim, Ratiopharm’s Ratiograstim, Hexai’s Filgrastim Hexal, and Hospira’s Nivestim, all approved between September 15, 2008 and June 8, 2010.Footnote 82

Neulasta (pegfilgrastim) is a second-generation injectable granulocyte colony stimulating factor approved in both the US and EU. Pegfilgrastim is distinguished from filgrastim agents primarily because of pegfilgrastim’s longer serum half-life, implying generally less frequent dosing than the filgrastims. Specifically, whereas filgrastim requires daily dosing, pegfilgrastim is administered only once per chemotherapy cycle.Footnote 83 Currently Neulasta is patent-protected in the US and EU. According to Grabowski (2013, Slide 5), while the earliest reported year of key US patent expiry is 2013 for Amgen’s Neupogen, for its Neulasta it is 2015.

Infliximab (Remicade)

The EMA’s Committee on Human Medicinal Products (CHMP) approved Hospira’s biosimilar application for infliximab (injection) on September 10, 2013 with Centocor’s (Johnson & Johnson) Remicade serving as the reference product. Infliximib is a monoclonal antibody, an immunomodulator distributed as a lyophilized powder for solution. In the EU it is approved for treating ankylosing spondylitis (a chronic inflammatory disease of unknown origin, first affecting the spine and adjacent structures and commonly progressing to eventual fusion of the involved jointsFootnote 84), psoriatic arthritis and psoriasis (see Table 1 in main text). In addition to these three indications, in the U.S. infliximab (Remicade) is approved by the FDA for treatment of Crohn disease, fistulizing Crohn disease, rheumatoic arthritis, and ulcerative colitis.Footnote 85 Infliximab is the first monoclonal antibody approved as a biosimilar in the EU. Like several other immunologic agents, infliximab has a risk of serious infections, since patients treated with infliximab are at an increased risk for developing serious infections that may lead to hospitalization or death. According to Drug Facts and Figures (2011, p. 2822), “Most patients who developed those infections were taking concomitant immunosuppressants such as methotrexate or corticosteroids.”

Since EMA approval of biosimilar infliximab (referenced to Remicade) occurred just several months ago, data on its uptake within EU countries are not yet available. Indeed, it is likely that Hospira has not yet obtained reimbursement approval from any of the EMA member countries.

Follitropin Alfa (Gonal-F)

The EMA’s Committee on Human Medicinal Products (CHMP) approved Teva’s biosimilar application for follitropin alfa (injection) on September 27, 2013 with Merck Serono’s Gonal-F serving as the reference product. Follitropin alfa is a human follicle stimulating hormone (FSH) distributed as a sterile, clear solution for subcutaneous injection. In the EU it is approved for treating an ovulation (the failure of the ovaries to release an egg during an ovulation cycleFootnote 86), stimulation of multifollicular development in women undergoing superovulation for assisted reproductive technologies (ART) such as in vitro fertilization (IVF), gemete intra-fallopian transfer and zygote intra-fallopian transfer, and for ovaleap in association with a luteisising hormone (LH) preparation for women with severe LH and FSH deficiency. It is also approved in the EU for the stimulation of spermatogenesis in men who have congenital or acquired hypogonadotropic hypogonadism with concomitant human chorionic gonadotropin (hCG) therapy.Footnote 87 In the U.S. follitropin alfa FDA approved indications are limited to an ovulation and ART treatments.Footnote 88 Follitropin alfa is the first fertility biosimilar approved in the EU.

Since EMA approval of biosimilar follitropin alfa (referenced to Gonal-F) occurred just several months ago, data on its uptake within EU countries are not yet available. Indeed, it is likely that Teva has not yet obtained reimbursement approval from any of the EMA member countries.

Rights and permissions

Reprints and permissions

Copyright information

© 2015 American Association of Pharmaceutical Scientists

About this chapter

Cite this chapter

Berndt, E.R., Trusheim, M.R. (2015). Biosimilar and Biobetter Scenarios for the US and Europe: What Should We Expect?. In: Rosenberg, A., Demeule, B. (eds) Biobetters. AAPS Advances in the Pharmaceutical Sciences Series, vol 19. Springer, New York, NY. https://doi.org/10.1007/978-1-4939-2543-8_15

Download citation

Publish with us

Policies and ethics