Skip to main content

Part of the book series: Advances in Biochemical Engineering/Biotechnology ((ABE,volume 165))

Abstract

Therapeutic proteins show a rapid market growth. The relatively young biotech industry already represents 20 % of the total global pharma market. The biotech industry environment has traditionally been fast-pasted and intellectually stimulated. Nowadays the top ten best selling drugs are dominated by monoclonal antibodies (mABs).

Despite mABs being the biggest medical breakthrough in the last 25 years, technical innovation does not stand still.

The goal remains to preserve the benefits of a conventional mAB (serum half-life and specificity) whilst further improving efficacy and safety and to open new and better avenues for treating patients, e.g., improving the potency of molecules, target binding, tissue penetration, tailored pharmacokinetics, and reduced adverse effects or immunogenicity.

The next generation of biopharmaceuticals can pose specific chemistry, manufacturing, and control (CMC) challenges. In contrast to conventional proteins, next-generation biopharmaceuticals often require lyophilization of the final drug product to ensure storage stability over shelf-life time. In addition, next-generation biopharmaceuticals require analytical methods that cover different ways of possible degradation patterns and pathways, and product development is a long way from being straight forward. The element of “prior knowledge” does not exist equally for most novel formats compared to antibodies, and thus the assessment of critical quality attributes (CQAs) and the definition of CQA assessment criteria and specifications is difficult, especially in early-stage development.

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

Access this chapter

Institutional subscriptions

References

  1. Miller KL, Lanthier M (2015) Regulatory watch: innovation in biologic new molecular entities: 1986-2014. Nat Rev Drug Discov 14(2):83–83

    CAS  PubMed  Google Scholar 

  2. Elvin JG, Couston RG, van der Walle CF (2013) Therapeutic antibodies: market considerations, disease targets and bioprocessing. Int J Pharm 440(1):83–98

    CAS  PubMed  Google Scholar 

  3. Milstein C (1999) The hybridoma revolution: an offshoot of basic research. Bioessays 21(11):966–973

    CAS  PubMed  Google Scholar 

  4. Smith GP, Petrenko VA (1997) Phage display. Chem Rev 97(2):391–410

    CAS  PubMed  Google Scholar 

  5. Riechmann L et al (1988) Reshaping human antibodies for therapy. Nature 332(6162):323–327

    CAS  PubMed  Google Scholar 

  6. Story CM (1994) A major histocompatibility complex class I-like Fc receptor cloned from human placenta: possible role in transfer of immunoglobulin G from mother to fetus. J Exp Med 180(6):2377–2381

    CAS  PubMed  Google Scholar 

  7. Jakovljevic MB (2014) Oncology monoclonal antibodies expenditure trends and reimbursement projections in the emerging Balkan market. Farmeconomia. Health Econ Ther Pathways 15(1):27–32

    Google Scholar 

  8. Carter PJ (2011) Introduction to current and future protein therapeutics: a protein engineering perspective. Exp Cell Res 317(9):1261–1269

    CAS  PubMed  Google Scholar 

  9. Saag KG et al (2008) American College of Rheumatology 2008 recommendations for the use of nonbiologic and biologic disease-modifying antirheumatic drugs in rheumatoid arthritis. Arthritis Care Res 59(6):762–784

    CAS  Google Scholar 

  10. Singh JA et al (2010) Biologics for rheumatoid arthritis: an overview of Cochrane reviews. Sao Paulo Med J 128(5):309–310

    PubMed  Google Scholar 

  11. Udpa N, Million RP (2015) Monoclonal antibody biosimilars. Nat Rev Drug Discov 15:13

    PubMed  Google Scholar 

  12. Zolot RS, Basu S, Million RP (2013) Antibody–drug conjugates. Nat Rev Drug Discov 12(4):259–260

    CAS  PubMed  Google Scholar 

  13. Ornes S (2013) Antibody–drug conjugates. Proc Natl Acad Sci U S A 110(34):13695–13695

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Alley SC, Okeley NM, Senter PD (2010) Antibody–drug conjugates: targeted drug delivery for cancer. Curr Opin Chem Biol 14(4):529–537

    CAS  PubMed  Google Scholar 

  15. Jain N et al (2015) Current ADC linker chemistry. Pharm Res 32(11):3526–3540

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Le LN et al (2012) Profiling antibody drug conjugate positional isomers: a system-of-equations approach. Anal Chem 84(17):7479–7486

    CAS  PubMed  Google Scholar 

  17. Beckley NS et al (2013) Investigation into temperature-induced aggregation of an antibody drug conjugate. Bioconjug Chem 24(10):1674–1683

    CAS  PubMed  Google Scholar 

  18. Singh SK, Luisi DL, Pak RH (2015) Antibody-drug conjugates: design, formulation and physicochemical stability. Pharm Res 32(11):3541–3571

    CAS  PubMed  Google Scholar 

  19. Junutula JR et al (2008) Site-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic index. Nat Biotechnol 26(8):925–932

    CAS  PubMed  Google Scholar 

  20. Shen B-Q et al (2012) Conjugation site modulates the in vivo stability and therapeutic activity of antibody-drug conjugates. Nat Biotechnol 30(2):184–189

    CAS  PubMed  Google Scholar 

  21. Axup JY et al (2012) Synthesis of site-specific antibody-drug conjugates using unnatural amino acids. Proc Natl Acad Sci U S A 109(40):16101–16106

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Senter PD, Sievers EL (2012) The discovery and development of brentuximab vedotin for use in relapsed Hodgkin lymphoma and systemic anaplastic large cell lymphoma. Nat Biotechnol 30(7):631–637

    CAS  PubMed  Google Scholar 

  23. Younes A, Yasothan U, Kirkpatrick P (2012) Brentuximab vedotin. Nat Rev Drug Discov 11(1):19–20

    CAS  PubMed  Google Scholar 

  24. Ricart AD (2011) Antibody-drug conjugates of calicheamicin derivative: gemtuzumab ozogamicin and inotuzumab ozogamicin. Clin Cancer Res 17(20):6417–6427

    CAS  PubMed  Google Scholar 

  25. Kim MT et al (2014) Statistical modeling of the drug load distribution on trastuzumab emtansine (Kadcyla), a lysine-linked antibody drug conjugate. Bioconjug Chem 25(7):1223–1232

    CAS  PubMed  Google Scholar 

  26. Chowdhury R, Ellis P (2014) Trastuzumab (Herceptin®) and Ado‐Trastuzumab Emtansine (Kadcyla®): treatments for HER2‐positive breast cancer. In: Handbook of therapeutic antibodies, pp 2041–2068

    Google Scholar 

  27. Cartwright H (2011) BMS/Ambrx deal highlights big pharma’s growing interest in protein drug engineering. PharmaDeals Rev 2011(10):1756

    Google Scholar 

  28. Agarwal P et al (2013) Hydrazino-Pictet-Spengler ligation as a biocompatible method for the generation of stable protein conjugates. Bioconjug Chem 24(6):846–851

    CAS  PubMed  Google Scholar 

  29. Beck A, Reichert JM (2011) Therapeutic Fc-fusion proteins and peptides as successful alternatives to antibodies. MAbs 3:415

    PubMed  PubMed Central  Google Scholar 

  30. Czajkowsky DM et al (2012) Fc-fusion proteins: new developments and future perspectives. EMBO Mol Med 4(10):1015–1028

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Kienast Y et al (2013) Ang-2-VEGF-A CrossMab, a novel bispecific human IgG1 antibody blocking VEGF-A and Ang-2 functions simultaneously, mediates potent antitumor, antiangiogenic, and antimetastatic efficacy. Clin Cancer Res 19(24):6730–6740

    CAS  PubMed  Google Scholar 

  32. Heier JS et al (2012) Intravitreal aflibercept (VEGF trap-eye) in wet age-related macular degeneration. Ophthalmology 119(12):2537–2548

    PubMed  Google Scholar 

  33. Matthews JE et al (2008) Pharmacodynamics, pharmacokinetics, safety, and tolerability of albiglutide, a long-acting glucagon-like peptide-1 mimetic, in patients with type 2 diabetes. J Clin Endocrinol Metabol 93(12):4810–4817

    CAS  Google Scholar 

  34. Herrington-Symes AP et al (2013) Antibody fragments: prolonging circulation half-life special issue-antibody research. Adv Biosci Biotechnol 4:689

    Google Scholar 

  35. Holliger P, Hudson PJ (2005) Engineered antibody fragments and the rise of single domains. Nat Biotechnol 23(9):1126–1136

    CAS  PubMed  Google Scholar 

  36. Manning MC et al (2010) Stability of protein pharmaceuticals: an update. Pharm Res 27(4):544–575

    PubMed  Google Scholar 

  37. Wang W et al (2007) Antibody structure, instability, and formulation. J Pharm Sci 96(1):1–26

    CAS  PubMed  Google Scholar 

  38. Mahler HC et al (2009) Protein aggregation: pathways, induction factors and analysis. J Pharm Sci 98(9):2909–2934

    CAS  PubMed  Google Scholar 

  39. Harris RJ et al (2001) Identification of multiple sources of charge heterogeneity in a recombinant antibody. J Chromatogr B Biomed Sci Appl 752(2):233–245

    CAS  PubMed  Google Scholar 

  40. Mahler HC et al (2010) Protein aggregation and particle formation: effects of formulation, interfaces, and drug product manufacturing operations. Aggregation Ther Proteins:301–331

    Google Scholar 

  41. Singh SK (2011) Impact of product-related factors on immunogenicity of biotherapeutics. J Pharm Sci 100(2):354–387

    CAS  PubMed  Google Scholar 

  42. Chi E et al (2003) Physical stability of proteins in aqueous solution: mechanism and driving forces in nonnative protein aggregation. Pharm Res 20(9):1325–1336

    CAS  PubMed  Google Scholar 

  43. Singh SK et al (2010) An industry perspective on the monitoring of subvisible particles as a quality attribute for protein therapeutics. J Pharm Sci 99(8):3302–3321

    CAS  PubMed  Google Scholar 

  44. Mahler HC, Borchard G, Luessen HL (eds) (2010) Protein pharmaceuticals: formulation, analytics and delivery. ECV-Editio Cantor-Verlag, Mainz

    Google Scholar 

  45. Jarasch A et al (2015) Developability assessment during the selection of novel therapeutic antibodies. J Pharm Sci 104(6):1885–1898

    CAS  PubMed  Google Scholar 

  46. Pikal MJ (2002) Freeze drying. Encyclopedia of Pharmaceutical Technology, Marcel Dekker, New York. 1299, p 1326

    Google Scholar 

  47. Wang W (1999) Instability, stabilization, and formulation of liquid protein pharmaceuticals. Int J Pharm 185(2):129–188

    CAS  PubMed  Google Scholar 

  48. Haag R (2004) Supramolecular drug-delivery systems based on polymeric core–shell architectures. Angew Chem Int Ed Engl 43(3):278–282

    CAS  PubMed  Google Scholar 

  49. Leriche G, Chisholm L, Wagner A (2012) Cleavable linkers in chemical biology. Bioorg Med Chem 20(2):571–582

    CAS  PubMed  Google Scholar 

  50. Ouyang J (2013) Drug-to-antibody ratio (DAR) and drug load distribution by hydrophobic interaction chromatography and reversed phase high-performance liquid chromatography. Methods Mol Biol 1045:275–283

    PubMed  Google Scholar 

  51. Pikal MJ, Shah S (1990) The collapse temperature in freeze drying: dependence on measurement methodology and rate of water removal from the glassy phase. Int J Pharm 62(2):165–186

    CAS  Google Scholar 

  52. Roy M et al (1991) The effects of formulation and moisture on the stability of a freeze-dried monoclonal antibody-vinca conjugate: a test of the WLF glass transition theory. Dev Biol Stand 74:323–339 discussion 340

    Google Scholar 

  53. Tang XC, Pikal MJ (2004) Design of freeze-drying processes for pharmaceuticals: practical advice. Pharm Res 21(2):191–200

    CAS  PubMed  Google Scholar 

  54. Murase N, Franks F (1989) Salt precipitation during the freeze-concentration of phosphate buffer solutions. Biophys Chem 34(3):293–300

    CAS  PubMed  Google Scholar 

  55. Bhatnagar BS, Bogner RH, Pikal MJ (2007) Protein stability during freezing: separation of stresses and mechanisms of protein stabilization. Pharm Dev Technol 12(5):505–523

    CAS  PubMed  Google Scholar 

  56. Rambhatla S et al (2004) Heat and mass transfer scale-up issues during freeze drying: II. Control and characterization of the degree of supercooling. AAPS PharmSciTech 5(4):54–62

    PubMed Central  Google Scholar 

  57. Geidobler R, Winter G (2013) Controlled ice nucleation in the field of freeze-drying: Fundamentals and technology review. Eur J Pharm Biopharm 85(2):214–222

    CAS  PubMed  Google Scholar 

  58. Fissore D, Pisano R, Barresi AA (2011) Advanced approach to build the design space for the primary drying of a pharmaceutical freeze-drying process. J Pharm Sci 100(11):4922–4933

    CAS  PubMed  Google Scholar 

  59. Pikal M (1985) Use of laboratory data in freeze drying process design: heat and mass transfer coefficients and the computer simulation of freeze drying. PDA J Pharm Sci Technol 39(3):115–139

    CAS  Google Scholar 

  60. Schneid SC et al (2015) Application of process analytical technology for monitoring freeze-drying of an amorphous protein formulation: use of complementary tools for real-time product temperature measurements and endpoint detection. J Pharm Sci 104(5):1741–1749

    CAS  PubMed  Google Scholar 

  61. Schneid SC et al (2011) Optimization of the secondary drying step in freeze drying using TDLAS technology. AAPS PharmSciTech 12(1):379–387

    PubMed  PubMed Central  Google Scholar 

  62. Pikal M et al (1990) The secondary drying stage of freeze drying: drying kinetics as a function of temperature and chamber pressure. Int J Pharm 60(3):203–207

    CAS  Google Scholar 

  63. Carpenter JF et al (1997) Rational design of stable lyophilized protein formulations: some practical advice. Pharm Res 14(8):969–975

    CAS  PubMed  Google Scholar 

  64. Wang W (2000) Lyophilization and development of solid protein pharmaceuticals. Int J Pharm 203(1):1–60

    CAS  PubMed  Google Scholar 

  65. Franks F (1998) Freeze-drying of bioproducts: putting principles into practice. Eur J Pharm Biopharm 45(3):221–229

    CAS  PubMed  Google Scholar 

  66. Prestrelski SJ et al (1993) Dehydration-induced conformational transitions in proteins and their inhibition by stabilizers. Biophys J 65(2):661–671

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Kolhe P, Amend E, Singh SK (2010) Impact of freezing on pH of buffered solutions and consequences for monoclonal antibody aggregation. Biotechnol Prog 26(3):727–733

    CAS  PubMed  Google Scholar 

  68. Yu A et al (1998) Pain perception following subcutaneous injections of citrate-buffered and phosphate-buffered epoetin alpha. Int J Artif Organs 21(6):341–343

    CAS  PubMed  Google Scholar 

  69. Carpenter J, Prestrelski S, Arakawa T (1993) Separation of freezing-and drying-induced denaturation of lyophilized proteins using stress-specific stabilization: I. Enzyme activity and calorimetric studies. Arch Biochem Biophys 303(2):456–464

    CAS  PubMed  Google Scholar 

  70. Glomb MA, Monnier VM (1995) Mechanism of protein modification by glyoxal and glycolaldehyde, reactive intermediates of the Maillard reaction. J Biol Chem 270(17):10017–10026

    CAS  PubMed  Google Scholar 

  71. Chang BS, Kendrick BS, Carpenter JF (1996) Surface-induced denaturation of proteins during freezing and its inhibition by surfactants. J Pharm Sci 85(12):1325–1330

    CAS  PubMed  Google Scholar 

  72. Hawe A et al (2009) Structural properties of monoclonal antibody aggregates induced by freeze–thawing and thermal stress. Eur J Pharm Sci 38(2):79–87

    CAS  PubMed  Google Scholar 

  73. Pikal-Cleland KA et al (2000) Protein denaturation during freezing and thawing in phosphate buffer systems: monomeric and tetrameric β-galactosidase. Arch Biochem Biophys 384(2):398–406

    CAS  PubMed  Google Scholar 

  74. Singh SK et al (2011) Frozen state storage instability of a monoclonal antibody: aggregation as a consequence of trehalose crystallization and protein unfolding. Pharm Res 28(4):873–885

    CAS  PubMed  Google Scholar 

  75. Gikanga B et al (2015) Mixing monoclonal antibody formulations using bottom-mounted mixers: impact of mechanism and design on drug product quality. PDA J Pharm Sci Technol 69(2):284–296

    CAS  PubMed  Google Scholar 

  76. Allmendinger A et al (2015) Sterile filtration of highly concentrated protein formulations: impact of protein concentration, formulation composition, and filter material. J Pharm Sci 104:3319–3329

    CAS  PubMed  Google Scholar 

  77. Tyagi AK et al (2009) IgG particle formation during filling pump operation: a case study of heterogeneous nucleation on stainless steel nanoparticles. J Pharm Sci 98(1):94–104

    CAS  PubMed  Google Scholar 

  78. Shieu W, Stauch OB, Maa Y-F (2015) Filling of high-concentration monoclonal antibody formulations into pre-filled syringes: investigating formulation-nozzle interactions to minimize nozzle clogging. PDA J Pharm Sci Technol 69(3):417–426

    CAS  PubMed  Google Scholar 

  79. Shieu W et al (2014) Filling of high-concentration monoclonal antibody formulations into pre-filled syringes: filling parameter investigation and optimization. PDA J Pharm Sci Technol 68(2):153–163

    PubMed  Google Scholar 

  80. Torosantucci R et al (2013) Identification of oxidation sites and covalent cross-links in metal catalyzed oxidized interferon beta-1a: potential implications for protein aggregation and immunogenicity. Mol Pharm 10(6):2311–2322

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Kishore RS et al (2011) The degradation of polysorbates 20 and 80 and its potential impact on the stability of biotherapeutics. Pharm Res 28(5):1194–1210

    CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Roman Mathaes .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2018 Springer International Publishing AG

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Mathaes, R., Mahler, HC. (2018). Next Generation Biopharmaceuticals: Product Development. In: Kiss, B., Gottschalk, U., Pohlscheidt, M. (eds) New Bioprocessing Strategies: Development and Manufacturing of Recombinant Antibodies and Proteins. Advances in Biochemical Engineering/Biotechnology, vol 165. Springer, Cham. https://doi.org/10.1007/10_2016_57

Download citation

Publish with us

Policies and ethics