Overview of Biosimilar Therapeutics

Overview of Biosimilar Therapeutics

C H A P T E R 5 Overview of Biosimilar Therapeutics Danuta J. Herzyk Merck Research Laboratories, Merck & Co., Inc., West Point, PA, USA INTRODUCTIO...

190KB Sizes 265 Downloads 281 Views

C H A P T E R

5 Overview of Biosimilar Therapeutics Danuta J. Herzyk Merck Research Laboratories, Merck & Co., Inc., West Point, PA, USA

INTRODUCTION As described in Section I of this book, biotechnology-derived pharmaceuticals represent a very broad group of medicines in terms of their structure and therapeutic indications. Also, there are fundamental differences between traditional, small-molecule pharmaceuticals and biological products. The physiochemical and structural properties of biologics are sensitive to changes in the manufacturing environment and/or processes, and because of the inherent variability in the biological systems (i.e., living cells) used in the manufacturing process, the resulting biologic product often consists of a mixture of protein isoforms. Because of the complexity of biopharmaceuticals, for many years it was believed their manufacturing could not be subject to generic competition. This view changed in the early 2000s in parallel with significantly increased knowledge and progress in biotechnological methods applied in biopharmaceutical manufacturing. At the same time greater public and governmental pressure on lower cost medicines and larger global access to novel therapeutics led to this evolutionary step in the biotechnology world. The primary focus on the discovery and development of novel biologics has been broadened to the development of copies of successful marketed biopharmaceuticals in a less expensive way. While it is widely accepted that in contrast to chemical drugs, generic versions of biologics are not attainable (because of the high complexity of their structure), the development and manufacturing of close copies of biopharmaceuticals is achievable, and the concept of “biosimilar” drugs has been introduced. The term “biosimilar” refers to a biological product that claims to be similar to a marketed biopharmaceutical (reference product) that has been approved in highly regulated markets, such as International Conference for Harmonization (ICH) regions (i.e. USA, Europe, Japan), for which patent and regulatory protection of data exclusivity have expired. There are other terms, such as “follow-on- biologics” and “subsequent entry biologics,” applied in different regions (USA and Canada, respectively) that refer to the same definition of biological products as “biosimilars.” In this chapter the term “biosimilar” has been adopted since it is the most broadly used term associated with copies of biological reference products. Nonclinical Development of Novel Biologics, Biosimilars, Vaccines and Specialty Biologics. http://dx.doi.org/10.1016/B978-0-12-394810-6.00005-8

141

© 2013 Elsevier Inc. All rights reserved.

142

5.  OVERVIEW OF BIOSIMILAR THERAPEUTICS

Development of biosimilar products encompasses great challenges on two fronts, the first related to the very complex nature of biopharmaceutical entities and the second linked to the multifaceted and highly protected manufacturing processes of these molecules. While information about any biopharmaceutical molecular structure becomes publicly known upon the receipt of marketing authorization, information about biopharmaceutical manufacturing and purification processes, specifications, and raw materials used is proprietary and not available in the public domain. Moreover, for many biopharmaceutical products patent protection is separate for the molecular structure and the formulation of the product, and patent for the formulation may last beyond the expiration of patent for the structure of the molecule. Biosimilar manufacturers do not have access to the innovator’s DNA clone or original cell bank and must engineer and clone a new cell line to produce a copy of the original product. Overall, biosimilar manufacturers face a certain “knowledge gap” regarding the development history and database with critical quality product attributes of the innovator’s product.

THE CONCEPT OF BIOSIMILARS In the past few decades most scientific discussions related to the development of novel biologics were focused on establishing optimal scientific and regulatory approaches to their safety evaluation, particularly in nonclinical and early clinical phases of the drug development process. These fruitful discussions and shared experience resulted in the generation of regulatory guidelines [1] and publications [2] that solidified the principles applied in the development of biopharmaceuticals. Since a large number of biological products have become very effective medicines and financially successful, recently the debate has been shifted toward the possibility of the development of biosimilar drugs. Clearly, there are two opposite positions within the biopharmaceutical industry and its representative organizations that influence the debate. The proponents state that the development and commercialization of biosimilars has become reality and these drugs can help address unmet medical needs by improving access to well-established therapeutic interventions by improving their affordability. However, this goal can only be achieved when an abbreviated, less costly development paradigm under a new regulatory framework is in place [3,4]. The opponents claim that the limited testing of biosimilar candidates, especially in the clinic, is insufficient to ensure the same safety and efficacy profile of biosimilars when compared to the clinically proven benefits of marketed reference products [5]. According to this view, a quite extensive nonclinical and clinical evaluation of biosimilar candidates should be required before their approval for marketing, which would maintain the high cost of the development and effectively prevent the emergence of biosimilars. From the scientific point view, the development of biosimilars involves reversed protein engineering of the reference product, generation of the best feasible copy of the reference product using updated biotechnology methods, and an extensive comparative characterization of the reference and the biosimilar candidate using state-of-the-art techniques. Given that at least a decade may have passed between the generation and testing of the original biologic molecule and the biosimilar, more advanced technology than what the innovator was able to use during the development of the original biopharmaceutical would likely be in place. Because of inevitable changes in both biotechnology processes and characterization methods

II.  DEVELOPMENT OF BIOSIMILARS

The Concept of Biosimilars

143

over time and the intention to decrease the cost of manufacturing, a biosimilar candidate cannot be an exact replica of the innovator product. Chemistry, Manufacturing, and Control (CMC) processes required in the development of biological drugs are equally fundamental for both novel biologics and biosimilars, and need to be conducted according to the existing regulatory guidelines and standards. Twelve ICH documents have been put in place to address quality and CMC processes involved in the characterization and manufacturing of biopharmaceuticals [6–17], reflecting both the complexity of these products and the high regulatory scrutiny surrounding their quality. One fundamental difference in the development of a novel biologic drug candidate versus a biosimilar candidate is the extent of nonclinical and clinical studies. For the original novel biologic, it must be demonstrated in multiple animal and human studies that it will have the intended pharmacological properties and a good safety profile in relationship to its biochemical and physical characteristics. As these characteristics have been already established, for a biosimilar candidate, deemed to be a satisfactory copy of the reference product based on results generated in the CMC space, the goal is to confirm in relevant but limited studies that the pharmacological activity, efficacy, and safety profile is highly similar to its original biologic counterpart. In order to achieve this goal, great efforts are being made to understand the relationship between recombinant protein structure and CMC processes as well as potential consequences of small changes in CMC on functional characteristics and clinical activity of biotechnology products. As such, the challenges in the development of biosimilars may actually lead to scientific advances and innovation in biotechnology. The development of biosimilars is difficult not only because of the structural complexity of biologic drugs but also because the original biologic products often undergo changes in their manufacturing processes over the course of a product life cycle (i.e. after obtaining the marketing authorization). Manufacturers make post-marketing changes for many different reasons including manufacturing capacity, improving purity, increasing product yield, or optimizing the cost of goods [18]. When such manufacturing process changes are made, a manufacturer is mandated to demonstrate that the product pre- and post-change has comparable “identity, strength, quality, purity, and potency” [19] and the safety and efficacy data derived from nonclinical and clinical testing during the development of the pre-change product are relevant to the post-change product. The process of assuring product consistency pre- and post-change has been referred to as a “comparability” evaluation. To separate this comparability process performed within the same manufacturer (innovator) from the exercise involved in comparing characteristics of a biosimilar candidate with its reference product by another manufacturer (biosimilar developer) who, unlike the innovator, does not possess historical knowledge of the manufacturing processes involved in making the original product, the term “similarity” is applied to the comparative evaluation of biosimilars. Technically, however, it is difficult to distinguish between bioanalytical “comparability” and “similarity” as they involve the same parameters to determine quality characteristics of two biological products when tested side-by-side. In fact, the demonstration of biosmilarity requires both analytical comparability and similarity evaluation. The former involves the comparison of multiple lots of the innovator product to establish a range of product variance. The latter involves the comparison of the biosimilar candidate to the innovator product to confirm that multiple quality attributes remain inside the variability brackets (between the lowest and the highest values) for multiple lots of both products.

II.  DEVELOPMENT OF BIOSIMILARS

144

5.  OVERVIEW OF BIOSIMILAR THERAPEUTICS

Biosimilarity studies performed in recent years have shown that different lots of marketed biopharmaceuticals (e.g. rituximab, entanercept) can substantially vary in some attributes, such as glycosylation patterns due to process changes implemented during post-marketing manufacturing [3]. However, the analytically detectable changes did not appear to have any impact on the therapeutic (safety and efficacy) attributes of these biological drugs. This observation suggests that some degree of structural differences in biologics may be functionally inconsequential and, therefore, acceptable. On the other hand, biologic product safety and efficacy, primarily affected by increased immunogenicity, can be altered in the post-marketing phase by undetectable or unforeseen factors related not only to manufacturing process changes but also to modifications in formulation, container closure systems as well as in-use stability profiles. Each of these factors can affect product characteristics, including immunogenic potential [20,21]. Notably, aggregation in biologic drugs has been implicated as a critical factor in the induction of an immunogenic reaction and potential autoimmunity. The most discussed example of unforeseen increased immunogenicity, likely related to a change in the product formulation and its closure system, resulting in antibody-mediated pure red cell aplasia is Eprex, a recombinant human erythropoietin [22]. This case indicates that small differences in certain attributes of biological products can occur, despite the existing manufacturer’s experience in well-controlled and seemingly understood manufacturing process, and can have a detrimental impact on safety of the product after its approval. It is postulated that if some variability in quality attributes exists for the original biologic product manufactured based on acquired in-depth knowledge of the process, the probability of introducing much greater variability into its biosimilar counterpart is undoubtedly higher. The burden to prove that this postulate may not necessarily be true, upon application of high-quality reverse protein engineering and bioprocess expertise [3], is on biosimilar developers. In summary, existing experience in the development and approval of novel biopharmaceuticals, including comparability of pre- and post-change evaluation, is highly related and applicable to the emerging experience in the development and approval of biosimilars. Based on current scientific knowledge in characterization of the complex structure of biopharmaceuticals, detection of a small structural difference alone may or may not be meaningful and a “totality of the evidence” approach has been introduced as the most appropriate for assessing biosimilars [23].

GENERAL CONSIDERATIONS FOR DEVELOPMENT OF BIOSIMILARS The development of biosimilars requires significant resource investment in establishing manufacturing processes (bioprocess) and analytical method development to enable production and comparative characterization of the molecular structure and quality attributes of the target reference product and its copy. The early phase of biosimilar development is more extensive than the early development of novel biologics because it involves extensive analytical characterization of two products—the innovator and the biosimilar candidate. It may also be quite costly because of the necessity of purchasing a large quantity of the reference product, often manufactured at different sites and marketed as multiple versions in different countries, to be used in comparative evaluation. Thus, CMC work on biosimilars requires not only the development of a consistent and well-controlled manufacturing process of the biosimilar

II.  DEVELOPMENT OF BIOSIMILARS

General Considerations for Development of Biosimilars

145

product itself but also extensive comparative side-by-side analyses of procured reference product lots and manufactured biosimilar candidate batches. Once the analytical characterization of the intended biosimilar candidate and the reference product has demonstrated a “highly similar quality profile” as described in the European Medicines Agency (EMA) guidance [24], the extent of nonclinical and clinical studies with the biosimilar candidate should be reduced compared to the development of novel biopharmaceuticals as the clinical efficacy and safety of the reference product is well understood. However, the challenge is to define “highly similar” in light of expected or detected structural variance between the reference product and its biosimilar, and to know “how similar is similar enough” as addressed by Kozlowski et al. [23]. The comparative nonclinical and clinical studies with a proposed biosimilar and the reference product should be designed to detect potential small differences and to minimize uncertainty around whether or not the detected differences could translate to any meaningful difference between the tested products in their clinical application. A biosimilarity assessment is typically performed in a stepwise manner as recommended by new regulatory guidelines described in Chapter 2. Comparison of the proposed biosimilar product and the reference product includes the evaluation of structure, function, animal pharmacokinetics (PK), pharmacodynamics (PD), and toxicity, human PKPD, safety, efficacy, and immunogenicity.

Comparative Nonclinical Studies The aim of comparative studies is to identify (qualitatively and quantitatively) potential differences in product attributes between the biosimilar candidate and reference product. In vitro tests Extensive structural and functional characterization is the first and most critical step in the development of biosimilars. This characterization requires the establishment of multiple in vitro assays and product-specific reagents as well as assay qualification and/or validation. Methods and model systems need to be tailored to a specific biologic product. In addition, the selected methods need to be demonstrated as capable of detecting small differences in all aspects of biochemical characterization of biologics. The conduct of comparative studies involves multiple batches of the biosimilar candidate and commercial lots of the innovator products. Using appropriate analytical methods with adequate sensitivity and specificity for structural characterization of the proteins, the comparisons of the biosimilar candidate and reference product should include: primary structures, such as amino acid sequence, higher order structures, including secondary, tertiary, and quaternary structure, post-translational modifications, such as glycosylation and phosphorylation, and other potential protein changes that can involve unintentional (e.g. deamidation and oxidation during storage) or intentional chemical modifications (e.g. PEGylation). Functional assays need to be designed to characterize pharmacologic activity of the biosimilar candidate in comparison to the reference product. These assays typically include cell-based assays (bioassays) and binding assays involving many different techniques (e.g. immunoassays) to evaluate potency of tested products. By nature, functional assays are highly variable and require careful optimization of testing conditions before they can be reliably used in comparative testing.

II.  DEVELOPMENT OF BIOSIMILARS

146

5.  OVERVIEW OF BIOSIMILAR THERAPEUTICS

If rigorous structural and functional comparisons in in vitro studies show minimal or no difference in these assays, the probability of achieving an adequate biosimilar candidate is high and the next step of in vivo evaluation is pursued. In vivo pharmacology, PK and PD studies Theoretically, a high degree of similarity between the biosimilar candidate and the reference product demonstrated by in vitro studies should allow for the reduction in the number of required in vivo tests. In practice, however, many animal studies are conducted for most biosimilar candidates to build a strong comparative database and maximize similarity information. Typically, at least one animal disease model is developed to conduct comparative pharmacology (efficacy) studies using multiple versions of the reference product and biosimilar candidate. For example, a neutropenic mouse model for the evaluation of granulocyte colony-stimulating factor activity, murine xenograft tumor models for comparison of the activity of anticancer monoclonal antibodies, and rheumatoid arthritis models in transgenic mice expressing human tumor necrosis factor (TNF) for the evaluation of TNF inhibitors have been used in the development of pertinent biosimilar candidates. Sometimes the animal models are newly established during the development of a biosimilar candidate because such models were not available at the time the innovator biologic was developed. Because of the comparative nature of these studies, they are rather costly as they require a large number of animals and the procurement of the innovator products. Similar to in vitro testing, if several versions of the reference product are marketed in different countries it is prudent to use multiple lots/versions of the reference product in the in vivo evaluation to determine the variability range of animal responses to this product. In addition to animal pharmacology experiments in disease models, similarity in PKPD characteristics between the biosimilar candidate and its reference product (multiple versions if applicable) is also assessed in normal healthy animals. In order to perform animal PKPD studies for comparative purposes, appropriate product-specific assays have to be developed and qualified by a biosimilar developer. Small differences in quality and/or in vitro functional characteristics between the reference product and the biosimilar candidate are typically detectable and should be expected. Small inconsistencies in animal pharmacology, PK and/or PD studies could also be observed, including some variation between multiple lots of the reference product. Therefore, the identified dissimilarities may or may not represent any meaningful differential response to the biosimilar candidate compared to the reference product and they are often confounded by natural inter-animal variability, including an immune response to a human biologic (i.e. immunogenicity of the tested products). When some differences between the biosimilar candidate and the reference product are observed (greater than the range of variability between reference lots) in PKPD animal studies, additional (larger) studies may be needed to understand the nature of the found variance. When animal testing does not reveal any significant difference between the biosimilar candidate and its reference product in side-by-side comparative studies, the confidence in developing of an adequate biosimilar product is increased. The most likely scenario (based on personal communication) in the development of biosimilars is that small CMC differences are detectable using in vitro assays (outside their variability range) while in vivo PKPD comparative studies do not show significant changes (beyond the natural or explainable variability) between the biosimilar candidate and the reference

II.  DEVELOPMENT OF BIOSIMILARS

General Considerations for Development of Biosimilars

147

product. This scenario is in contrast to the position presented in the past that in vitro analytical methods used in characterization of biologics are less sensitive in detection of potential differences than the whole host (in vivo). A shift in thinking about the reliance on in vitro testing in comparative studies is evident in new regulatory guidelines for biosimilars [25,26]. Toxicology studies To address any uncertainty about translation of small CMC differences to the safety profile of a biosimilar candidate, a toxicology study is typically expected. Since the safety profile of the reference product is well understood in humans, the extent of the nonclinical toxicology program in the development of a biosimilar to support its clinical testing and registration should be abbreviated when compared to the toxicology program performed during the development of the reference product. At the same time, a toxicology program for a biosimilar candidate needs to be guided by information generated for the pertinent reference product and utilize product-specific approaches. Typically, one repeated dose toxicology study, comparative in nature in the same animal species as that used by the innovator, is conducted. The study design, including dose levels, frequency, and duration of dosing should be driven by results of the pivotal toxicity study performed by the innovator. Although a toxicity study with a biosimilar candidate should be based on product-specific characteristics, experience in toxicological sciences indicates that generally a repeat-dose study of a minimum duration of one-month in which the biosimilar and the reference product are tested side by side is considered sufficient. Such a study is quite resource intensive as it requires many groups of animals and the procurement of large amounts of the reference product. Unlike pharmacology and PKPD studies that include multiple lots/versions of the reference product, in toxicology studies testing one version of the reference product is considered sufficient based on guidelines for animal use and/ or data generated from comparative PKPD assessments. Regarding the biosimilar candidate material to be tested in a comparative toxicology study, it is highly desirable to use drug product from the optimized manufacturing process that is representative of the material to be tested in pivotal clinical trials designed to evaluate the safety profile of the biosimilar candidate. Typically, no other toxicology studies (i.e. safety pharmacology, chronic toxicity, reproductive and developmental toxicity) are expected for biosimilars unless there is a specific cause for concern. Additional consideration should be given when no toxicity was detected in animal studies performed by the innovator during the development of the reference product (e.g. TNFα inhibitors) and where no differences between the biosimilar candidate and the reference product are identified upon nonclinical comparative characterization using both in vitro (according to the current standards of quality testing) and in vivo (including PKPD) studies. It is highly debatable that a comparative toxicology study should be conducted in such a scenario. The regulatory guidelines for biosimilars [25,26] indicate that the lack of a comparative (presumably in the situation described above) or any toxicology study with a biosimilar candidate (e.g. due to lack of relevant toxicology species) can be justified by sponsors developing biosimilars. Limited experience to date (based on personal communication) suggests that comparative toxicology studies do not detect any meaningful differences (beyond the natural or explainable variability) between biosimilar candidates and their reference products even when some degree of “dissimilarity” in other nonclinical characterization has been observed.

II.  DEVELOPMENT OF BIOSIMILARS

148

5.  OVERVIEW OF BIOSIMILAR THERAPEUTICS

Comparative Clinical Studies Clinical development of biosimilars, although abbreviated, is still fairly extensive. The clinical program should always include at least one human comparative study using the biosimilar and the reference product side by side. The number and type of additional studies with the biosimilar candidate to confirm the expected efficacy and safety in patients can vary based on the characteristics of the reference products. The clinical studies provide the ultimate data to support the evidence of similarity between the biosimilar candidate and the reference product. The comparative nature of clinical studies with a biosimilar is focused on bioequivalence and requires new clinical trial expertise compared to conventional clinical trials, in which efficacy and safety are measured against placebo controls. In addition, most clinical trial investigators, primarily researchers at university hospitals, are much more interested in studying novel drug candidates than evaluating copies of the existing medicines, as the latter does not satisfy their pursuit of advancing clinical sciences. In some cases, where competition for patient enrollment in certain diseases is very high, clinical investigators are deeply critical of trials with biosimilars, viewing them as “stealing” patients from “real” clinical trials. This point of view can be countered, however, by the fact that in a comparative trial with the biosimilar candidate and the reference product, all patients benefit from being treated by a pharmacologically active and efficacious agent with no chance of being exposed to a placebo control, which does not provide any therapeutic effect. On top of such disputes, the new regulatory guidelines for biosimilars, either finalized or drafted [25–27], outline stringent requirements for clinical trials that should include a dose–response comparison between the biosimilar and the reference product, testing the most sensitive patient population to detect potential differences, and long-term monitoring of immunogenicity both prior to registration and during the post-marketing period. Also, the guidelines recommend using drug product from a large-scale commercial manufacturing process that is fully defined prior to the start of clinical trials. While such a rigorous evaluation would minimize any uncertainties associated with the approval of biosimilars, it will also minimize incentives and justification for investments in the development of biosimilars in light of only small reduction in costs compared to the development of new biologic entities. Because of this conundrum, the regulatory requirements for clinical development of biosimilars are still evolving and incomplete.

BIOSIMILAR CANDIDATES BASED ON MODALITY AND THERAPEUTIC CLASS Biopharmaceuticals include many different types of molecular constructs and can be grouped in three major categories: (1) therapeutic proteins (e.g. enzymes, hormones, cytokines); (2) monoclonal antibodies (mAbs), and modifications thereof, such as, mAbs conjugated to chemicals or immunoglobulin fragments (e.g. Fab); and (3) fusion proteins involving the Fc portion of immunoglobulin linked to other recombinant proteins. Lists of marketed biologics in these three categories are presented in Tables 5.1 (A, B, C) and 5.2 (A, B, C). Another categorization of biopharmaceuticals can be based on therapeutic area and disease indication. The major classes of biopharmaceuticals include drugs for treatment of cancer,

II.  DEVELOPMENT OF BIOSIMILARS

TABLE 5.1  Biopharmaceuticals Approved for Marketing (US) in 1983–2005 Generic name (brand name) Year

Target /ligand

Indication

Therapeutic area

Interferon α-2b (Intron A) 1983

Interferon alfa-2b

Leukemias, melanoma, hepatitis C, hepatitis B

Oncology, infectious diseases

Interferon α-2a (Roferon A) 1984

Interferon alfa-2a

Leukemias, hepatitis C

Oncology, infectious diseases

Somatropin (Humatrope) 1987

Growth hormone

Growth hormone deficiency

Endocrinology

Interferon α-N3 (Alferon) 1989

Interferon alfa-N3

Genital warts

Various

Filgrastin (Neupogen) 1991

Granulocyte colony-stimulating factor Neutropenia (G-CSF)

Oncology and others

Sargramostim (Leukine) 1991

Granulocyte macrophage colonystimulating factor (GM-CSF)

Oncology

Aldesleukin (Proleukin) 1992

Interleukin-2 (IL-2) receptor on T and Metastatic renal cell carcinoma NK cells

Oncology

Dornase alpha (Pulmozyne) 1993

DNase

Cystic fibrosis

Pulmonary diseases

Epoetin alpha (Epogen/Procrit) 1993

Erythropoietin

Anemia

Oncology and others

Imiglucerase (Cerezyme) 1994

Glucocerebrosidase

Gaucher’s disease

Enzyme replacement therapy

Asparaginase (Elspar) 1994

l-Asparaginase

Acute lymphocytic leukemia

Oncology

Pegasparagase (Oncaspar) 1994

Pegylated l-asparaginase

Acute lymphoblastic leukemia

Oncology

Somatropine (Genotropine) 1995

Growth hormone

Growth hormone deficiency

Endocrinology

Alteplase (Activase) 1996

Tissue plasminogen activator

Restoration of function of central venous access devices

Cardiovascular diseases

A. THERAPEUTIC PROTEINS

Interferon beta-1a

Multiple sclerosis

Inflammation/autoimmunity

Mutation of tissue plasminogen activator

Acute myocardial infarction

Cardiovascular diseases

Follitropin (Follistim) 1997

Follicle-stimulating hormone

Infertility

Reproductive system

Interferon alphacon (Intergen) 1997

Interferon type 1

Hepatitis C

Infectious diseases

149

Interferon β-1a (Avonex) 1996 Reteplase (Retavase) 1996

Biosimilar Candidates Based on Modality and Therapeutic Class

II.  DEVELOPMENT OF BIOSIMILARS

Chemotherapy-related acute myelogenous leukemia

Continued

Generic name (brand name) Year

Target /ligand

150

TABLE 5.1  Biopharmaceuticals Approved for Marketing (US) in 1983–2005  (cont’d)

Indication

Therapeutic area

Interleukin-11 (IL-11)

Thrombocytopenia in chemotherapy

Oncology

Glucagon (Glucagen) 1998

Glucagon hormone

Hypoglycemic reactions in diabetic patients

Endocrinology

Interferon γ-1b (Actimune) 1999

Interferon gamma-1b

Infections with chronic granulomatous diseases, malignant osteoporosis

Infectious diseases, oncology

Insulin glargine (Lantus) 2000

Long-acting insulin

Diabetes

Endocrinology

Insulin aspart (Novolog) 2000

Rapid-acting insulin

Diabetes

Endocrinology

Tenecteplase (Tnkase) 2000

Tissue plasminogen activator

Myocardial infarction

Cardiovascular diseases

Darbepoetin alpha (Aranesp) 2001

Erythropoietin

Anemia

Oncology and others

Anakinra (Kineret) 2001

IL-1 receptor antagonist

Rheumatoid arthritis

Inflammation/autoimmunity

Drotrecogin alpha (Xigris) 2001

Activated protein C

Sepsis

Infectious diseases

PegInterferon α-2b (Peg-Intron) 2001

Interferon alfa-2b, pegylated

Hepatitis C

Infectious diseases

Interferon β-1a (Rebif) 2002

Interferon beta-1a, pegylated

Multiple sclerosis

Inflammation/autoimmunity

PegInterferon α-2a (Pegasys) 2002

Interferon alfa-2a, pegylated

Hepatitis C, hepatitis B

Infectious diseases

Pegfilgrastim (Neulasta) 2002

Pegylated G-CSF

Neutropenia

Oncology and others

Rasburicase (Elitek) 2002

Urate oxidase

Cancer-related hyperuricemia

Oncology

Teriparatide (Forteo) 2002

Parathyroid hormone

Osteoperosis

Bone diseases

Agalisdase beta (Fabrazyme) 2003

Alpha galactosidase

Fabry’s disease

Enzyme replacement therapy

Interferon β-1b (Betaseron) 2003

Interferon beta-1b

Multiple sclerosis

Inflammation/autoimmunity

Laronidase (Adlurazyme) 2003

α-l-iduronidase

Mucopolysaccharidosis I

Enzyme replacement therapy

Insulin glulisine (Apidra) 2004

Rapid-acting insulin

Diabetes

Endocrinology

Palifermin (Kepivance) 2004

Keratinocytes growth factor

Oral mucotitis in cancer

Oncology

5.  OVERVIEW OF BIOSIMILAR THERAPEUTICS

II.  DEVELOPMENT OF BIOSIMILARS

Oprelvekin (Neumega) 1997

Insulin (Levemir) 2005

Long-acting basal insulin

Diabetes

Endocrinology

Galsulfase (Naglazyme) 2005

N-Acetylgalactosamine 4-sulfase

Mucopolysaccharidosis VI

Enzyme replacement therapy

Mecasermin, (Increlex) 2005

Insulin growth factor-1 (IGF-1)

Growth failure secondary to severe primary IGF-1 deficiency

Endocrinology

Muromomab (Orthoclone OKT3) 1992

T-cell surface protein CD3 antagonist (murine antibody)

Rejection of transplanted organs

Organ transplant

Abciximab (Reopro) 1997

Platelet glycoprotein GpIIbIIIa receptor (Fab fragment) antagonist

Acute blood clot complications

Cardiovascular diseases

Daclizumab (Zenapax) 1997

IL-2 receptor (IL-2R, CD25) antagonist Rejection of transplanted organs

Organ transplant

Rituximab (Rituxan) 1997

B-cell surface protein CD20 antagonist

Non-Hodgkin’s lymphoma, rheumatoid arthritis

Oncology, inflammation/ autoimmunity

Trastuzumab (Herceptin) 1997

Human epidermal growth factor receptor 2 (HER2) antagonist

Breast cancer

Oncology

Infliximab (Remicade) 1998

Tumor necrosis factor (TNF) alpha antagonist

Rheumatoid arthritis, Crohn’s disease, psoriatic arthritis, ankylosing spondylitis, psoriaris

Inflammation/autoimmunity

Basiliximab (Simulect) 1998

IL-2R alpha chain (CD25) antagonist

Organ transplant

Immunology

Palivizumab (Synagis) 1998

F protein of respiratory syncytial virus (RSV) antagonist

Prophylaxis of serious lower respiratory tract disease

Infectious diseases

Gemtuzumab ozogamicin (Mylotarg) 2000

Anti-CD33 conjugated with calicheamicin

Acute myeloid leukemia

Oncology

Alemtuzumab (Campath) 2001

B-cell surface protein CD52 antagonist

B-cell chronic lymphocytic leukemia

Oncology

Adalimumab (Humira) 2002

TNF alpha antagonist

Rheumatoid arthritis, Crohn’s disease Inflammation/autoimmunity

Ibritumomab tiuxetan (Zevalin) 2002

Anti-CD20 linked to radioactive yttrium-90

Non-Hodgkin’s lymphoma

Oncology

131I-Tositumomab

Anti-CD20 linked to radioactive iodine-131

Non-Hodgkin’s lymphoma, chronic lymphocytic leukemia

Oncology

B. MONOCLONAL ANTIBODIES

151

Continued

Biosimilar Candidates Based on Modality and Therapeutic Class

II.  DEVELOPMENT OF BIOSIMILARS

(Bexxar) 2003

152

TABLE 5.1  Biopharmaceuticals Approved for Marketing (US) in 1983–2005  (cont’d)

Target /ligand

Indication

Therapeutic area

II.  DEVELOPMENT OF BIOSIMILARS

Efalizumab (Raptiva) 2003

Integrin lymphocyte functionassociated antigen-1 (CD11a) antagonist

Psoriasis

Inflammation/autoimmunity

Omalizumab (Xolair) 2003

IgE antagonist

Asthma

Pulmonary diseases

Bevacizumab (Avastin) 2004

Vascular endothelial growth factor (VEGF) antagonist

Colorectal cancer

Oncology

Cetuximab (Erbitux) 2004

Epidermal growth factor receptor (EGFR) antagonist

Colorectal cancer

Oncology

Etanercept (Enbrel) 1998

TNFalpha (TNFR:Fc) antagonist

Rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, psoriaris

Inflammation/autoimmunity

Alefacept (Amevive) 2003

Leukocyte function antigen-3 (LFA3:Fc) antagonist

Psoriasis

Inflammation/autoimmunity

C. FUSION PROTEINS

5.  OVERVIEW OF BIOSIMILAR THERAPEUTICS

Generic name (brand name) Year

153

Biosimilar Candidates Based on Modality and Therapeutic Class

TABLE 5.2  Biopharmaceuticals Approved for Marketing (US) after 2005 Generic name (brand name) Date

Target/ligand

Indication

Therapeutic area

A. THERAPEUTIC PROTEINS Alglucosidase α (Myozyme) 2006

Alglucosidase alpha

Pompe diseases

Enzyme replacement therapy

Insulin (Exubera) 2006

Glucose metabolism

Type 1 or type 2 diabetes

Endocrinology

Idursulfase (Elaprase) 2006

Idursulfase

Hunter syndrome mucopolysaccharidosis II

Enzyme replacement therapy

Somatropine (Omnitrope) 2006

Sustained release of growth hormone

Growth failure

Endocrinology

PegEpo (Mircera) 2007

Methoxy polyethylene glycol-epoetin beta

Anemia associated with chronic renal failure

Various

Ecallantide (Kalbitor) 2009

Plasma kallikrein inhibitor

Hereditary angioedema

Genetic disorders

Pegloticase (Krystexxa) 2010

Pegylated uricase

Refractory chronic gout

Inflammation/ autoimmunity

B. MONOCLONAL ANTIBODIES Natalizumab (Tysabri) 2006

Integrin alpha4beta1 antagonist

Multiple sclerosis

Inflammation/ autoimmunity

Panitumumab (V Ectibix) 2006

EGFR1 antagonist

Colorectal cancer

Oncology

Ranibizumab (Lucentis) 2006

VEGF-R antagonist (antibody fragment)

Macular degeneration, agerelated, wet; Retinal vein occlusion; Edema

Ophthalmology

Eculizumab (Soliris) 2007

Complement factor C5 inhibitor

Hemolytic anemia, hemolytic uremic syndrome

Hematology

Certolizumab pegol (Cimzia) 2008

TNFalpha antagonist (pegylated Fab fragment)

Crohn’s disease, rheumatoid arthritis

Inflammation/ autoimmunity

Canakinumab (Ilaris) 2009

IL-1beta antagonist

Muckle–Wells syndrome, familial cold autoinflammatory syndrome, neonatal onset multisystem inflammatory disease

Inflammation/ autoimmunity

Golimumab (Simponi) 2009

TNFalpha antagonist

Rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis

Inflammation/ autoimmunity

Ofatumumab (Arzerra) 2009

B-cell CD20 antagonist

Chronic lymphocytic leukemia

Oncology

Ustekinumab (Stelara) 2009

IL-12/IL-23 antagonist

Psoriasis

Inflammation/ autoimmunity Continued

II.  DEVELOPMENT OF BIOSIMILARS

154

5.  OVERVIEW OF BIOSIMILAR THERAPEUTICS

TABLE 5.2  Biopharmaceuticals Approved for Marketing (US) after 2005 (cont’d) Generic name (brand name) Date

Target/ligand

Indication

Therapeutic area

Denosumab (Xgeva, Prolia) 2010

Receptor activator of nuclear factor kappa-B ligand (RANKL) antagonist

Osteoporosis, bone regeneration (cancer-related bone loss)

Bone diseases

Tocilizumab (Actemra) 2010

IL-6R antagonist

Castleman’s disease, rheumatoid arthritis, juvenile arthritis

Inflammation/ autoimmunity

Belimumab (Benlysta) 2011

BLyS (B-cell survival factor) antagonist

Systemic lupus erythematosus, rheumatoid arthritis

Inflammation/ autoimmunity

Brentuximab Vendotin (Adcetris) 2011

Anti-CD30 (cell surface protein) conjugated to monomethyl auristatin

Refractory Hodgkin lymphoma

Oncology

Ipilimumab (Yervoy) 2011

CTLA-4 (extracellular domain) antagonist

Melanoma

Oncology

Abatacept (Orencia) 2006

T-cell CD80/CD86 co-stimulation (CTLA4:Fc) inhibitor

Rheumatoid arthritis

Inflammation/ autoimmunity

Rilonacept (Arcalyst) 2008

IL-1 receptor (IL-1R:Fc) antagonist

Cryopyrin-associated periodic syndromes

Inflammation/ autoimmunity

Romiplastim (Nplate) 2008

Thrombopoietin (TPO:Fc) receptor agonist

Chronic immune thrombocytopenia

Inflammation/ autoimmunity

Aflibercept (Eylea) 2011

VEGF trap (specific domain of VEGFR1/ VEGFR2:Fc)_

Neovascular (wet) age-related Ophthalmology macular degeneration

Belatacept (Nulojix) 2011

Cytotoxic T-cell inhibitor via CD28: CD80/CB86 interaction (extracellular CTLA-4:Fc)

Kidney transplant rejection

C. FUSION PROTEINS

Organ transplant

inflammatory, and autoimmune diseases, metabolic disorders, enzyme and hormone deficiencies, and others (Tables 5.1 and 5.2). The tables include biologic products approved for marketing in the US as new biological entities (NBE) in chronological order, allowing for the appreciation of evolutionary changes in the range and types of biotechnology-derived medicines. Some of the changes are focused on improving convenience of biologic drug administration (e.g. by pegylation of recombinant proteins, their half-life has been prolonged and frequency of parenteral administration reduced). Other modifications are focused on expanding disease indications by changing target binding specificity through additional engineering

II.  DEVELOPMENT OF BIOSIMILARS

Summary

155

of biologic constructs. Such modifications could be made by the same innovator or a competitor, but in each case a new application for approval as a NBE had to be filed and marketing authorization received. A number of marketed biologics, with the expired or imminent loss of patent protection, have been selected as targets for biosimilars based on high revenues. In contrast to the desired and deliberate modifications to improve biologic products that are already effective therapeutics, biosimilars can not intentionally be modified. The first wave of biosimilars approved in EU markets represents a class of therapeutic proteins and include growth hormone (somatotropin), erythropoietin (epoetin), and granulocyte colony-stimulating factor (filgrastim). Based on publication of specific guidance for biosimilars to insulin and interferon [28,29] one can assume that biosimilar candidates to marketed insulin products and interferon products may be in development. The “blockbuster” biologics include several mAbs to treat inflammatory diseases and cancer, and these drugs are expected to be targets for biosimilars upon their patent expiration. The mAbs that may face competition from the development of their biosimilars include TNFblocking products (e.g. adalimumab), anti-CD20 antibody (rituximab), anti-HER2 antibody (trastuzumab), and anti-VEGF (bavacizumab). Particulars related to the nonclinical development of biosimilars, mainly to selected marketed therapeutic proteins, are discussed in Chapter 3.

SUMMARY Global interest in access to lower cost biopharmaceuticals in the form of biosimilars aligns with the concept of generic medications for chemical pharmaceuticals. However, based on experience to date, development of biosimilars poses much greater challenges not only due to demanding requirements in clinical development, but also uncertainty in manufacturing and marketing capabilities. The development costs of a biosimilar, while lower than the cost of developing a novel biopharmaceutical, are still high. Manufacturing capabilities that require sophisticated facilities, technologies, and processes have proven to be elevated barriers to developing biosimilars. Interactions with physicians and healthcare providers require new resources in educational processes both prior to conduct of clinical trials and after obtaining marketing authorization. Sales teams need to acquire deeper medical and technical knowledge to explain why biosimilars are not generic substitutions of their original biopharmaceutical counterparts and yet should be trusted as equally safe and efficacious medicines. Likewise, investment in market strategies to meet required post-marketing surveillance is very significant. Lastly, slowly shaping regulatory framework, especially in the US, makes the investments quite risky. Nonetheless, growing interest in biosimilars in emerging markets, including India, China, and Brazil, where health authorities have published or drafted their own guidelines, will likely increase investments in biosimilars in those regions. In conclusion, the value proposition of biosimilars is complex and has to be addressed by the full range of stakeholders, including manufacturers, regulators, physicians, payers, and patients. If development, registration, and marketing of biosimilars become broadly successful, it will likely happen over the long term.

II.  DEVELOPMENT OF BIOSIMILARS

156

5.  OVERVIEW OF BIOSIMILAR THERAPEUTICS

References [1] ICH. ICH S6(R1) Preclinical Safety Evaluation of Biotechnologically-Derived Pharmaceuticals. www.ich.org/ products/guidelines/safety/article/safety-guidelines.html. Fed Reg 2012;77(97):29665–6; May 18, 2012. [2] Cavagnaro Joy A, editor. Preclinical Safety Evaluation of Biopharmaceuticals: A Science-Based Approach to Facilitating clinical Trials. Hoboken, NJ: John Wiley & Sons; 2008. [3] McCamish M, Woollett G. The state of the art in the development of biosimilars. Clin Pharmacol Ther 2011;343:1–3. [4] Schneider CK, Kalinke U. Toward biosimilar monoclonal antibodies. Nat Biotechnol 2008;26:985–90. [5] Davis GC, Beals JM, Johnson C, Mayer MH, Meiklejohn BI, Mitak BH, et al. Recommendations regarding technical standars for follow-on biologics: Comparability, similarity, interchangeability. Commentary. Curr Med Res Opin 2009;25:1655–61. [6] ICH. ICH Q2(R1) Validation of Analytical Procedures. Text and Methodology. CPMP/ICH281/95. www.ich. org/products/guidelines/quality/article/quality-guidelines.html; 1996. [7]  ICH. ICH Q3A(R2) Impurities in New Drug Substances. CPMP/ICH/142/95. www.ich.org/products/­ guidelines/quality/article/quality-guidelines.html; 2006. [8] ICH. ICH Q5A(R1) Viral Safety Evaluation of Biotechnology Products Derived from Cell Lines of Human or Animal Origin. PMSB/ELD Notification No. 329. www.ich.org/products/guidelines/quality/article/qualityguidelines.html; 1999. [9] ICH. ICH Q5B Quality of Biotechnological Products: Analysis of the Expression Construct in Cells Used for Production of r-DNA Derived Protein Products. CPMP/ICH/139/95. www.ich.org/products/guidelines/ quality/article/quality-guidelines.html; 1995. [10] ICH. ICH Q5C Stability Testing of Biotechnological/Biological Products. CPMP/ICH/138/95. www.ich.org/ products/guidelines/quality/article/quality-guidelines.html; 1995. [11] ICH. ICH Q5D Quality of Biotechnological/Biological Products: Derivation and 662 Characterization of Cell Substrates Used for Production of Biotechnological/Biological Products. CPMP/ICH/294/95Q5D. www.ich. org/products/guidelines/quality/article/quality-guidelines.html; 1997. [12] ICH. ICH Q5E. Comparability of Biotechnological/Biological Products Subject to Changes in their Manufacturing Process. www.ich.org/products/guidelines/quality/article/quality-guidelines.html; 2004. [13] ICH. ICH Q6B Specifications: Test Procedures and Acceptance Criteria for 669 Biotechnological/Biological Products. CPMP/ICH/365/96. www.ich.org/products/guidelines/quality/article/quality-guidelines.html; 1999. [14]  ICH. ICH Q7 Good Manufacturing Practice Guidance for Active Pharmaceutical Ingredients. CPMP/ ICH/4106/00. www.ich.org/products/guidelines/quality/article/quality-guidelines.html; 2000. [15] ICH. ICH Q8(R2) Pharmaceutical Development 674 675 19. CHMP/ICH/167068/0417. www.ich.org/­products/ guidelines/quality/article/quality-guidelines.html; 2009. [16] ICH. ICH Q9 Quality Risk Management. EXT/24235/2006. www.ich.org/products/guidelines/quality/ article/quality-guidelines.html; 2005. [17] ICH. ICH Q10 Pharmaceutical Quality System. CHMP/ICH/214732/04. www.ich.org/products/guidelines/ quality/article/quality-guidelines.html; 2008. [18] Lewis RM. Demonstration of comparability of a lincensed product after a manufacturing change. In: Cavagnaro Joy A, editor. Preclinical Safety Evaluation of Biopharmaceuticals: A Science-Based Approach to Facilitating Clinical Trials. Hoboken, NJ, 8: John Wiley & Sons; 2008. p. 161–78. [19] US FDA. FDA Guidance Concerning Demonstration of Comparability of Human Biological Products, Including Therapeutic Biotechnology-derived Products. www.fda.gov/cder/Guidance/compare.htm; 1996. [20] Hermeling S, Crommelin DJ, Schellekens H, Jiskoot W. Structure-immunogenicity relationships of therapeutic proteins. Pharm Res 2004;21:897–903. [21] Chirino AJ. Minimizing the immunogenicity of protein therapeutics. Drug Discov Today 2004;9:82–90. [22] Schellekens H, Crommelin DJ. Equivocal role of micelles in Eprex adverse events. Nat Biotechnol 2003;21: 1255–62. [23] Kozlowski S, Woodcock J, Midhun K, Behrman Sherman R. Developing the nation’s biosimilars program. N Engl J Med 2011;363:385–8.

II.  DEVELOPMENT OF BIOSIMILARS

References

157

[24] EMA. Guideline on Similar Biological Medicinal Products containing Biotechnology-derived Proteins as Active Substance. Quality Issues. London: 22 February 2006 EMEA/CHMP/BWP/49348/2005; 2006. http:// www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2009/09/WC500003953.pdf. [25] CHMP. CHMP Guideline on Similar Biological Medicinal Products containing Biotechnology-derived Proteins as Active Substance: Nonclinical and Clinical Issues. EMEA/CHMP/BMWP/42832/2005. www.ema.europa. eu/docs/en_GB/document_library/Scientific_guideline/2009/09/WC500003920.pdf; 2006. [26] FDA. FDA Scientific Considerations in Demonstrating Biosimilarity to a Reference Product. Draft Guidance. www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM291128.pdf; 2012. [27]  EMA. Guideline on Similar Biological Medicinal Products containing Monoclonal Antibodies—DRAFT. ­Adoption by CHMP for release for consultation, 18 November. www.ema.europa.eu/docs/en_GB/document_ library/Scientific_guideline/2010/11/WC500099361.pdf; 2010. [28] EMEA. Annex to Guideline on Similar Biological Medicinal Products Containing Biotechnology-derived Proteins as Drug Substance—Nonclinical and Clinical Issues containing Recombinant Human Insulin. EMEA/ CHMP/32775/2005. http://www.ema.europa.eu/ema/index.jsp?curl=pages/regulation/general/general_ content_000408.jsp; 2005. [29] EMEA. Guideline on Similar Biological Medicinal Products containing Recombinant Interferon alpha. EMEA/ CHMP/BMWP/102046/2006. www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2009/ 09/WC500003930.pdf; 2006.

II.  DEVELOPMENT OF BIOSIMILARS