biosimilars: science to market

82
i MSc Biotechnology, Bioprocessing And Business Management 2010-2011 1055902 Supervisor: Dr. Neil Porter Word Count: 15,169 A dissertation submitted in part fulfillment of the Degree of MSc. Biotechnology, Bio processing and Business Management, University of Warwick, September 2011 BIOSIMILARS: SCIENCE TO MARKET

Upload: priyesh-waghmare

Post on 12-Apr-2017

5.981 views

Category:

Health & Medicine


0 download

TRANSCRIPT

Page 1: BIOSIMILARS: SCIENCE TO MARKET

i

MSc Biotechnology, Bioprocessing

And Business Management

2010-2011

1055902

Supervisor: Dr. Neil Porter

Word Count: 15,169

A dissertation submitted in part fulfillment of the Degree of MSc. Biotechnology, Bio processing and Business

Management, University of Warwick, September 2011

BIOSIMILARS: SCIENCE TO MARKET

Page 2: BIOSIMILARS: SCIENCE TO MARKET

i

Acknowledgement

I would like to take this opportunity to thank my supervisor Dr. Neil Porter, for his invaluable

insight and guidance throughout my dissertation.

I would like to thank Dr. Crawford Dow, Dr. Steve Hicks and Dr. Charlotte Moonan for their

assistance and Adrienne Davis for her continuous support and encouragement throughout the

year.

Finally, I would like to thank my parents and friends, without whom this piece of thesis would

not have been possible.

Page 3: BIOSIMILARS: SCIENCE TO MARKET

ii

TABLE OF CONTENTS

List of Tables ............................................................................................................................................ v

List of Figures: .......................................................................................................................................... vi

Executive Summary................................................................................................................................ viii

1. Introduction: ........................................................................................................................................ 1

1.1 Biopharmaceuticals: ....................................................................................................................... 1

1.2 Biopharmaceutical Market: ............................................................................................................. 4

2. Biosimilars: .......................................................................................................................................... 6

2.1 Terminology disputes: .................................................................................................................... 7

2.2 Differences between Biosimilars & Generic Drugs: .......................................................................... 8

2.2.1 Product differences: ............................................................................................................... 10

2.2.2 Manufacturing differences: .................................................................................................... 10

2.3 Manufacturing Process: ................................................................................................................ 13

2.3.1 Challenges: ............................................................................................................................. 13

2.3.2 Selection of platform: ............................................................................................................. 15

2.3.3 Purification: ............................................................................................................................ 16

2.3.4 Formulation: .......................................................................................................................... 16

3. Biosimilars legislation & Regulations: ................................................................................................. 18

3.1 Regulatory framework Europe: ..................................................................................................... 19

3.1.1 Product specific guidelines: .................................................................................................... 24

3.1.2 Immunogenicity: .................................................................................................................... 26

3.1.3 Extrapolation:......................................................................................................................... 27

3.2 EU Biosimilar approval process: .................................................................................................... 28

3.2.1 Common technical document: ................................................................................................ 28

3.3 United States Regulatory framework: ........................................................................................... 30

Page 4: BIOSIMILARS: SCIENCE TO MARKET

iii

3.4 Global Landscape: ......................................................................................................................... 31

4. Case Study-Biosimilar Insulin: ............................................................................................................. 33

4.1 Manufacturing: ............................................................................................................................. 33

4.2 EMEA requirements: ..................................................................................................................... 34

4.3 Marvel’s Insulin rejection: ............................................................................................................. 35

4.3.1 Quality Aspects: ..................................................................................................................... 36

4.3.2 Non clinical aspects: ............................................................................................................... 36

4.3.3 Clinical Aspects:...................................................................................................................... 36

5. Market Analysis: ................................................................................................................................ 37

5.1 Biosimilars On market: .................................................................................................................. 37

Table 5.2: Unsuccessful biosimilar applications in the EU. Source: Greer, F.M., 2011 .............................. 39

5.2 Market Size & Growth: ................................................................................................................. 40

5.3 Market potential:.......................................................................................................................... 40

5.4 Regional market analysis: ............................................................................................................. 42

5.5 Biologic class market analysis: ...................................................................................................... 43

5.6 Market Opportunities: .................................................................................................................. 45

5.6.1 Patent expiry: ......................................................................................................................... 47

5.7 Market share: ............................................................................................................................... 54

5.8 Sales: ............................................................................................................................................ 56

5.9 market Drivers: ............................................................................................................................. 56

5.9.1 Cost Savings (Global health care): ........................................................................................... 56

6. Issues & Challenges: ........................................................................................................................... 60

6.1 Cost of Product development: ...................................................................................................... 60

6.1.1 US region ............................................................................................................................... 61

6.2 Manufacturing Facility: ................................................................................................................. 61

Page 5: BIOSIMILARS: SCIENCE TO MARKET

iv

6.3 Substitution: ................................................................................................................................. 62

6.4 Market exclusivity: ........................................................................................................................ 62

6.5 Innovator Strategies: .................................................................................................................... 64

6.6 Profitability of biosimilars: ............................................................................................................ 66

6.7 Marketing: .................................................................................................................................... 67

7. Conclusion: ........................................................................................................................................ 68

References: ............................................................................................................................................ 71

Page 6: BIOSIMILARS: SCIENCE TO MARKET

v

LIST OF TABLES

Table 1.1: Comparison of the size of the chemical and biological medicine 2

Table 1.2: Biopharmaceutical market, estimated value and forecast 2009-2015 in US$billion

5

Table 2.1: Biosimilars Terminology. 7

Table 2.2: Small molecule generics v/s biosimilars. 9

Table 2.3: Definitions of biological and chemical pharmaceuticals. 9

Table 2.4: comparison of generics, biosimilars & biologics. 11

Table 2.5: Biopharmaceutical processing with prokaryotic and eukaryotic expression systems.

15

Table 3.1. Format of the dossier- modules of the CTD. 29

Table 5.1: Biosimilars approved by the EU. 38

Table 5.2: Unsuccessful biosimilar applications in the EU. 39

Table 5.3: Total World Biosimilar Market Potential 2006-2013 41

Table 5.4: World Biosimilar Market Potential by Region 2006-2013 43

Table 5.6: The world market potential for Biosimilars by Biological Class (EPO, G-CSF, insulin, Interfereon, alpha, others) 2006-2013.

44

Table 5.7: Estimates of treatment cost per patient of selected biopharmaceuticals. Source: Crandall, 2009.

47

Table 5.8: Blockbuster biological drugs set to lose patent protection per year through 2015. Source: Emmerich, R. (2010)

49

Table 5.9: Interferons on market and patent expiries. Source: 50

Table 5.10: Multiple sclerosis drugs on the market and patent expiries. 52

Table 5.11: Recombinant insulin products on the market and patent expiries 53

Table 5.12: Bbiosimilar companies sales and market share 55

Table 6.1: Interferons on market and patent expiries 65

Page 7: BIOSIMILARS: SCIENCE TO MARKET

vi

LIST OF FIGURES:

Fig 1.1: Differences in complexity (biotech’s interferon)-a protein naturally produced in

our body versus Traditional AsprinSource ………………… 3

Fig 1.2: Evolution of the biologics market 2009-2015. 5

Fig 2.1: Biologic from production to drug use 14

Fig 3.1: Regulatory guidelines. 19

Fig 3.2: Market exclusivity. 25

Fig 4.1: Post fermentation steps in manufacturing process 34

Fig 5.1: World Biosimilar Market potential by region 2006-2013, products with currently

expired patents. 42

Fig 5.2: Expected Biosimilar market split in 2015 45

Fig 5.3: Forecast of the global biosimilar market value in $billion: 2008-12 46

Fig 5.4: Number and value of biological drugs set to lose patent protection per year

through 2015 48

Fig 5.6: Predicted market share of multiple sclerosis drugs (2007-2017) 51

Fig 5.7: Estimated patent expiry dates of selected proteins 54

Fig 5.8. Market Share of biosimilars in the off patent biologics market. 55

Fig 6.1: Patent protection and market exclusivity for top biologics losing patent protection prior to 2018

64

Page 8: BIOSIMILARS: SCIENCE TO MARKET

vii

ABBREVIATIONS

EMEA – European Medicine Agency

FDA – Food and Drug Administration

EBE - European Biopharmaceutical Enterprises

EU – European Union

ICH- International Conference on Harmonization

CHMP-Committee For Medicinal Products For Human Use

BLA- Biologic License Application

DNA-Deoxyribo Nucleic Acid

CMC- Chemistry Manufacturing and Controls

CTD- Common Technical Document

Page 9: BIOSIMILARS: SCIENCE TO MARKET

viii

EXECUTIVE SUMMARY

According to the definition, biosimilars are different versions of existing branded

biologics, which have received legal approval and which gain access to the market after the

demonstration of pre-clinical and clinical data proving their similarity to the reference product.

Due to their complex structure and nature as well as their complicated manufacturing

process biosimilars have become the subject of rigid regulatory frameworks currently in the

European Union and to be followed by the rest of the world.

The aim of this dissertation is to offer a wide spectrum view of biosimilars in general but

also in comparison to traditional generic chemical drugs.

In order to do this, an overview of the current regulatory frameworks focusing on EU and

US will be presented in relation to the manufacturing process and subsequently the approval

process.

Following this, an analysis of the market of biosimilars is offered addressing issues such

as market opportunities and drivers as well as the challenges faced.

Page 10: BIOSIMILARS: SCIENCE TO MARKET

1

1. INTRODUCTION:

The first generation of the biopharmaceuticals which are manufactured by the use of

recombinant technologies were launched in the 1980s and most of these products have either

already lost patent protection or are about to lose patent protection in the near future.

Biopharmaceuticals presently demand premium pricing due to various factors such as high cost

of manufacturing, superior safety and efficacy profiles and limited competition from other

biopharmaceutical companies. Since 1982 the global biopharmaceutical market has developed

significantly and was estimated to be worth $125 billion in 2010 (Greer, 2011).

Significant market opportunities for generic companies are provided by the expiry of the

patents first generation of biopharmaceutical/biotechnological products. Second-entry (follow-

on) biopharmaceutical/biotechnological products have a more complex route to the market as

compared to the generic versions of chemically-synthesized active ingredients. The different

terminology that is used to describe "biologically similar drugs" indicates the complexities in this

area. The generic industry tends to regard the biological similar drugs as the biogenerics, but the

research based industry argues that it is not possible to replicate precisely the biological process

for large molecules therefore, due to the nature of their production process; the generic of the

biopharmaceutical cannot exist (Marchant, 2007).

1.1 BIOPHARMACEUTICALS:

The biological medicines (biologic pharmaceuticals or biologics or biopharmaceuticals)

are the medicines which are produced using a living system or organism (EuropaBio 2005). The

division of drugs that are generated from biological sources and which include gene therapy,

vaccines, antibodies and other therapeutic products derived through biotechnology are called

as biologics or biopharmaceuticals (Wang, 2011).

Biopharmaceuticals are also considered as any substance used for the treatment or

management of diseases or injuries and is produced by natural organisms or recombinant

techniques consisting of proteins or other products derived from living organisms (Crandall,

Page 11: BIOSIMILARS: SCIENCE TO MARKET

2

2009). Using biotechnology, biopharmaceuticals are produced, which are medical drugs.

Biopharmaceuticals are proteins which include antibodies, nucleic acids (DNA, RNA or antisense

oligonucleotides) which are used for therapeutic or in vivo diagnostic purposes and are

produced by means other than direct extraction from a native (non-engineered) biological

source. Through a distinctive process biopharmaceuticals are produced, where various types of

bioreactors are used in which the microbial cells are cultured to produce proteins (Pandey, R. K.

et al., 2011).

The first biopharmaceutical product approved for therapeutic use was recombinant

human insulin (rHI), which also goes by the trade name Humulin. Humulin was developed by

Genentech and marketed by Eli Lilly & Co. in 1982.

The chemical medicines are usually organic molecules whose molecular structure can be

unfailingly assessed and they are produced by a defined chemical pathway (Fox, 2010). In

laboratory the chemical medicines are defined by simple analytical methods. The conventional

chemical medicines are different in various ways to the biological medicines. One of the

apparent differences is the size of the biopharmaceuticals; the molecules of the

biopharmaceutical are much larger, have more complex spatial structures and are to a great

extent heterogeneous than the small molecules which make up chemical medicines (Table 1.1).

Table 1.1: Comparison of the size of the chemical and biological medicines. Source: EuropaBio,2005

Page 12: BIOSIMILARS: SCIENCE TO MARKET

3

This makes it intricate to characterize biopharmaceuticals in a conventional way by

analyzing their individual components as is done for chemical medicines. A biopharmaceutical

product is molecule which is typically a protein with a complicated three dimensional structure

consisting of chain of hundreds of amino acids. Due the large size (Fig 1.1) and structure of the

molecules, the biopharmaceuticals are administered in injection form, whereas the chemical

medicines with small molecules come in pill form.

.

Fig 1.1: Differences in complexity (biotech’s interferon)-a protein naturally produced in our body versus Traditional AsprinSource: EuropaBio (2005) Biological and Biosimilar Medicines.

The productions conditions must be strictly controlled for the manufacturing of

biopharmaceuticals as they are very sensitive to the production processes. There is an

occurrence of complex post-translational modifications such as glycosylation and pegylation to

the protein, so even the small change in the manufacturing process could have a major impact

on biological activity. If compared in the terms of production quality tests, there are over 2000

production quality tests for the manufacture of a biological drug and only an average of 200

required for small molecule drugs (Pandey et al, 2011).

Page 13: BIOSIMILARS: SCIENCE TO MARKET

4

1.2 BIOPHARMACEUTICAL MARKET:

The biopharmaceuticals represent one of the most dynamic and potential segments of

the pharmaceutical sector and it has rapidly expanded over the past few years with

compounded growth rates which are beyond double digit figures, which are greater than the

performance of the overall pharmaceutical market (Taylor, 2009). In the field of biomedicine,

the biopharmaceuticals are well established and they have opened new avenues of therapy

options specifically in disease areas where earlier there were no therapies, or only insufficient

therapies were available (Kresse, 2009).

Since the early 1980s biopharmaceuticals have been a rising part of the pharmaceutical

sector. Biopharmaceuticals is one of the rapidly growing sectors in pharmaceutical industry,

growing at an average rate of 18-20% since 2007 (Crandall, 2009). There are many

biopharmaceuticals in the approval pipeline and it was projected that in 2010 for the market

place, 50% of drugs will be the result of biotechnology. There are some 165 biopharmaceutical

products which have gained approval. The total sales of recombinant protein-based drugs were

$54.5 billion in 2007 and in 2012 the sales are estimated to increase to $75.8 billion (Kresse,

2009).

Worldwide there are more than 400 new biopharmaceuticals under development or in

clinical trials and it has been recently estimated that biopharmaceutical sales will expand by 15-

20% annually in the future (Horikawa et.al, 2009). The biopharma market overall is forecasted to

grow at nearly 7% CAGR through to 2015 (Table 1.2), with MAbs (Monoclonal Antibodies)

showing higher growth of 9% (Evers, 2010). The biopharmaceutical market majorly comprises

of monoclonal antibodies, therapeutic proteins and vaccines. In terms of market size,

therapeutic proteins are the leaders (Fig 1.2), but Monoclonal Antibodies (mAbs) are the fastest

growing sector. MAbs (Monoclonal Antibodies) represents three quarters of the biologic market

and expected to dominate. Vaccines will have a steady growth rate and will hold their market

share. Therapeutic proteins are estimated to grow steadily but their growth rate will slightly

decline as compared to other product groups (Fig 1.2) (Evers, 2010).

Page 14: BIOSIMILARS: SCIENCE TO MARKET

5

Fig 1.2: Evolution of the biologics market 2009-2015. Source: Evers, P. (2010) The Future of the Biologicals Market.

Table 1.2: Biopharmaceutical market, estimated value and forecast 2009-2015 in US$billion

Source: Evers, P. (2010) The Future of the Biologicals Market.

Proteins Vaccines Mabs

52%

15%

33%38%

16%

46%

2009-$117bn

2015-$170bn

Page 15: BIOSIMILARS: SCIENCE TO MARKET

6

2. BIOSIMILARS:

The patent protection for most of the first- generation biopharmaceuticals began to

expire in 2004, opening the door to the so called ‘biosimilars’. A biosimilar is a medicine that is

similar but not identical to a biological medicine that has already been authorized (the

‘biological reference medicine’) (Zuniga & Calvo, 2009).

The biosimilars are also called follow-on biologics (FOB) or Subsequent Entry Biologics

which refer to the “generic” version of biologics or biopharmaceutical products that are

produced and sold on the market after the patents on the innovator’s biologics are expired.

However, the nomenclature of biosimilars is not universal (Wang, 2011).

Many definitions have been provided for “Biosimilars” by various authors. The

breakdown of the term “Biosimilars” can be done for the better understanding of the concept.

They are “biological medicinal products” which as the name suggests are similar to the

approved biological medicinal products in respect to quality, safety and efficacy. These

approved products are reference novel products which are already licensed and marketed. After

the reference product has lost patent protection and data/market exclusivity the independent

applicant can launch the biosimilar product after the approval. For the authorization of the

biosimilar product for marketing the applicant of the biosimilar producer or developer should

follow the procedure of regulations proving the similarity with the reference product. The

complex biological products are difficult to characterize completely, therefore the focus of the

“biosimilar” approach is generally on highly purified products consisting recombinant proteins

as the active pharmaceutical ingredient. According to the (Kresse, 2009) the approach is not

applicable to products which are derived from blood or plasma, immunologicals and other

upcoming therapies like gene or cell therapies. But the regulation bodies are prepared to accept

additional classes of compounds like polysaccharides such as low-molecular weight heparins.

Page 16: BIOSIMILARS: SCIENCE TO MARKET

7

2.1 TERMINOLOGY DISPUTES:

There has been extreme confusion among the regulatory bodies and countries about the

terminology which could be applied to the biopharmaceuticals/biologics that could be probably

accesible generically due to loss of patent protection and market exclusivity of the original

therapeutic protein (Crandall, 2009).

The complexity of the biopharmaceutical industry and the science behind it leads to

significant controversies with reference to definitions, terminology and issues related to

biopharmaceuticals in terms of products, technologies, companies. Biopharmaceutical are

complex medicines as compared to the small molecule chemical drugs as they are

manufactured by the usage of living organisms. Biopharmaceuticals possess complex nature,

size and complexity therefore they usually cannot be technically classified to the same extent as

the conventional chemical drugs (Taylor, 2009). As it is complicated to provide a concise

definition for a biopharmaceutical, it is particularly tricky to define a generic biopharmaceutical

given the complexity of the products derived from biotechnology. Table 2.1 illustrates the

different names which are used in various regions of the world to describe generic

biopharmaceuticals.

Table 2.1: Biosimilars Terminology. Source: Taylor P., 2009.

Page 17: BIOSIMILARS: SCIENCE TO MARKET

8

Most of the things associated with the concept of the biosimilars is controversial, even

the language related to the products. The term “biogenerics” is preferred by GPhA (Generic

Pharmaceutical Association), the generic industry trade group, as it indicates the possibility of

interchangeability and also improves the view of the public of generics as being as safe and

effective as the original product. Contrary to this belief the innovator companies approach is

different and use the term “follow –on biologics” (FOB). Different countries and regulatory

bodies of those countries make use of different terminology for generic biopharmaceuticals.

The European Union has the most established regulatory system for generic biopharmaceuticals

called EMA and this system makes use of the term “biosimilars.” The United States has switched

from the term “follow on protein product” to “follow-on biologic” to cover different kinds of

biologic product. Follow-on biologic is also considered as an umbrella term which covers both

biosimilars (i.e. products not having potential to substitute reference product) and biogenerics

(i.e. products having potential to substitute reference product) (Clark, 2009). At initial phases

most products have no proof of interchangeability and the European Union has an established

regulatory pathway as compared to the rest of the world which uses the term “biosimilar”

which will obliviously influence the regulatory system of United States and other countries.

2.2 DIFFERENCES BETWEEN BIOSIMILARS & GENERIC DRUGS:

Generic Medicines are the medicines which contain active substances whose safety and

efficacy are well established. Generic medicines must demonstrate that same dose of the

generic and reference product behave in the body in the exact same way which determines the

bioequivalence of the generic drug with that of the reference product (Zuniga & Calvo, 2009).

Quality in terms of the controls and standards for all manufacturing, preparation and processing

of the product should be maintained by the generic drug at the same standard to the reference

product. The generic drugs are generally considered as interchangeable with the reference

product because they are therapeutically equivalent to the reference product. The market

application procedure is relatively simple for generic medicines as there is no requirement of

results of clinical trials or the results of non clinical data like toxicological and pharmacological

tests. On the contrary it is completely opposite in the case of similar biological medicinal

Page 18: BIOSIMILARS: SCIENCE TO MARKET

9

products (biosimilars) whose development procedure is complicated like all the

biopharmaceutical products. For the biosimilar products the generic approach is not applicable

due to factors such as the unique manufacturing process for each product and complexity of the

products derived through biotechnology (Table 2.2) (Zuniga & Calvo, 2009). Table 2.3 provides

the definitions of the generic drug, biopharmaceutical and biosimilars.

Table 2.2: Small molecule generics v/s biosimilars. Source: Chen, 2009.

Table 2.3: Definitions of biological and chemical pharmaceuticals. Source: Crommelin et al., 2005.

Page 19: BIOSIMILARS: SCIENCE TO MARKET

10

2.2.1 PRODUCT DIFFERENCES:

The small molecule drugs called as chemical drugs due to their nature are able to

characterize chemically and this facilitates the generic manufacturers to evade the effort and

additional cost associated with clinical and non clinical evaluation and thereby proving their

product to be bioequivalent to the originator. The requirement of the accurate three

dimensional structures is necessary for the biological activity of the biopharmaceuticals, as this

structure helps in the interaction of the biopharmaceutical with other molecules like receptors

on cell surfaces, binding proteins and nucleic acids. During drug development there are various

profiles which should be fulfilled such as pharmacokinetic and pharmacodynamic profiles,

Clinical safety and efficacy profile all of which are influenced by the three dimensional structure

of biopharmaceutical, by the degree and location of its glycosylation sites, by its isoform profile

and by the degree of aggregation. The biosimilar product in order to prove equivalent to the

originator product has to have all these characteristics in addition to the primary (chemical)

structure to be identical to the original product and this makes the biosimilar different from

chemical generics which can be fully described by its chemical structure (Crommelin, 2005).

2.2.2 MANUFACTURING DIFFERENCES:

The manufacturing of the low molecular weight pharmaceuticals (chemical drugs) is

done by the sequence of controlled and conventional chemical reactions of the recognized

chemical reagents. Contrary to the chemical drugs, the biopharmaceuticals are manufactured

or produced by the harvesting of the proteins from the living cells which often results in the

additional secretion of various other substances along with the protein of interest (Table 2.4).

There is a general misunderstanding that biopharmaceutical product manufacturing is the

simple process of inserting the gene of interest in an appropriate cell line. However

biopharmaceutical manufacturing is a complex process which requires the attention of various

critical factors such as complex size and the three dimensional structures of biopharmaceuticals,

the unpredictable nature of the biological reactions with respect to chemical reactions, various

Page 20: BIOSIMILARS: SCIENCE TO MARKET

11

secondary modification processes such as glycosylation, and potential chances of denaturation,

aggregation and degradation (Crommelin et al., 2005).

Table 2.4: comparison of generics, biosimilars & biologics. Source: Accenture (2009)

There are several stages involved in the production of the biopharmaceuticals which could

affect the final product.

Page 21: BIOSIMILARS: SCIENCE TO MARKET

12

The characteristic of the protein product is determined by the selection of the host cell and

the sequence of genes that codes for the desired protein.

A master cell bank is established for protein production by extensive cell screening and

selection process and two master cell banks are never precisely the same

During the fermentation process the fermentor, components of the culture medium and

physical conditions at which the cells are cultured on the large scale affects the protein

which is produced and it also affects behavior properties in the body.

Purification is the critical step in manufacturing as various related proteins, DNA and other

purities are produced along with the protein of interest any change in the process could

affect the purity of the product.

A range of analytical techniques is used to examine different characteristics of the protein

product. Even the advanced analytical tools are not sufficient to analyze product

characteristic that may change the clinical safety and efficacy. On the other hand by using

only few analytical techniques the low molecular weight compounds can be completely

characterized in terms of structure.

The product should be stored cautiously due to the fact that if not stored in optimal

conditions, the product will lose its integrity.

The manufacturing of the biopharmaceutical is a more complex process as compared to the

small chemical synthetic drugs and to make exact reproduction of the innovator biologic

molecule is almost impossible (Greer, 2011). Every stage of the biopharmaceutical

manufacturing process is critical because the slightest change in the process can have

substantial change in the product and its efficacy in patients. The innovator faces a challenge to

maintain consistent batch to batch of biologic product in spite of possessing the patent

information and years of experience with the manufacturing process. Biosimilars cannot be

considered as regular generics as it is highly unlikely to manufacture a “copy” version of

biological products using the manufacturing process which is considerably different from the

innovators manufacturing process (Greer, 2011).

Page 22: BIOSIMILARS: SCIENCE TO MARKET

13

2.3 MANUFACTURING PROCESS:

Biosimilar development constitutes of three important steps such as Chemistry

Manufacturing and Controls (CMC), preclinical and clinical trials. The traditional generic drugs

are approved under an abbreviated pathway through the Hatch-Waxman Act of 1984. This

pathway is not applicable to the biosimilars as they come under biologics which are governed by

different laws and regulations. The ICH Common Technical Document (CTD) has a module 3 on

quality which describes the CMC requirements for the biosimilars. The European Union follows

the CTD format for submission of application and it will be used by the U.S., when the regulatory

pathway is put into place.

2.3.1 CHALLENGES:

To develop a biosimilar from scratch is a relatively tough task as the biosimilar

developers have no access to the proprietary information of the innovators product’s

manufacturing process or specification of the product. Generally biosimilars development has

to go through a series of steps such as the authorized marketed biologic product should be first

recognized by the biosimilar developer and used as the reference biologic product or innovators

product. The second step is to then carry out a thorough characterization of the reference

product. The manufacturing process of the biosimilar is developed from the data which is

generated from the characterization of the reference product. This data will also be utilized for

the comparability exercise which is performed to prove the bioequivalence between the

biosimilar and reference product. Due to the lack of access to the innovators manufacturing

process the biosimilar product is manufactured from entirely different and new process as

compared to the innovator manufacturing process. The new process developed by biosimilars

manufacturers may use and carry out production process in different culture system and

equipments like fermentor (Chen, 2009).

The challenges faced by the biosimilar as well as the innovator manufactures are the

same when it comes to the biopharmaceutical/ biologics production. Specifically there are two

challenges: Firstly there should be a robust manufacturing process which produces a consistent

Page 23: BIOSIMILARS: SCIENCE TO MARKET

14

product which has to be tested to be same in the preclinical and clinical studies. The second

challenge is to maintain product reproducibility in terms of scale up of the process with same

site or when manufacturing occurs at different sites. These two challenges are never easily met

by the manufacturers for instance the studies on innovators epoietin alpha with that of the

epoietin which is manufactured outside the U.S. and European Union have differences in terms

of purity, efficacy and biological activity. This example indicates that slight change in the

manufacturing process will result in the change in the biological activity of the product (Sharma,

2007). Fig 2.1 illustrates the flow from drug production to the administration.

Fig 2.1: Biologic from production to drug use

Source: Chen, B., 2009

Page 24: BIOSIMILARS: SCIENCE TO MARKET

15

2.3.2 SELECTION OF PLATFORM:

The development of the CMC is initiated by the selection of the production platform or

the expression system (Table 2.5). Depending on the type of protein to be manufactured an

appropriate technology is selected. The yeast expression system along with batch fermentation

is selected for the production of small peptides and proteins. The mammalian expression

system is selected as it provides the higher yields and reliable purification results and used for

the production of the monoclonal antibodies, complex proteins which contains disulphide

bonds and require glycosylation. Several stages in manufacturing are crucial and have influence

on the properties of the end product. Therefore after the selection and establishment of the cell

line which is crucial, a specific DNA sequence which codes for protein of interest is inserted

(Chen, 2009). Then extensive cell screening and appropriate methods are used to form a master

cell bank, so high levels of sterility and identity of the cells could be maintained. Throughout

the manufacturing process the cells are cultured under definite conditions so as to get

optimized results in terms of production and secretion of protein of interest. The structural

characteristics of the proteins are decided by the culture conditions in the mammalian cell

system and in the bacterial system they are later defined in the purification process (Sharma,

2007).

Table 2.5: Biopharmaceutical processing with prokaryotic and eukaryotic expression systems. Source: Sharma, 2007

Page 25: BIOSIMILARS: SCIENCE TO MARKET

16

2.3.3 PURIFICATION:

Impurities are unacceptable in the biologics manufacturing as impurities in the final

product could have clinical consequences. The process of purification has to get rid of all the

impurities which include cell proteins by host cells, DNA contamination, medium components,

viruses and other by products without causing any damage to the protein of interest. There

should be proper selection of the desired protein forms with suitable glycosylation and removal

of damaged forms in the purification process. Biosimilar manufacturers do not have access to

the purification process information of the innovator which leads to the differences in the purity

and protein structure. It is important for the biosimilar manufacturers to compromise on the

yield when it comes to purity as the purity leads to safety and efficacy status which is the

ultimate aim of the biosimilar. For instance in the erythropoietin purification, for the optimum

biological activity only the isoforms which are highly glycosylated are selected (Sharma, 2007).

2.3.4 FORMULATION:

The therapeutic performance and conformational stability of the biologic drug is

associated with composition of the formulation and choice of the container. These factors are

responsible for the protein degradation and aggregation. After going through composition of

formulation then sterile filtration and filling into the final container the final product is formed.

Components of the formulation consist of basic buffer which is for proper pH control and salt

which provides the isotonic adjustment. In order to avoid proteins from being absorbed to the

surface of containers or hydrophobic surfaces, surfactants are used in the formulation. In the

end container and closure reliability should be thoroughly checked for sterility. Biologics are the

heterogeneous mixtures which are not pure substances. Biologic products are produced under

the current Good Manufacturing Practice guidelines and to make sure they are produced

according to the predefined specifications there are various assays which could check the purity

and authenticity of the product. The biologics are sensitive to environmental changes such as

temperature, storage, handling and sunlight. For instance insulin vials exposed to temperature

Page 26: BIOSIMILARS: SCIENCE TO MARKET

17

more than 40C results in transformation of the product and increased chances of

immunogenicity and loss of biological activity (Sharma, 2007).

Page 27: BIOSIMILARS: SCIENCE TO MARKET

18

3. BIOSIMILARS LEGISLATION & REGULATIONS:

The regulatory bodies have recognized the fact that the manufacturing process is critical

in the production of biopharmaceuticals and variations in the process could lead to significant

differences in product characteristics which cannot be fully determined by the analytical

characterization. So the manufacturing process is considered and made part of the

determination of the product quality along with other testing protocols carried to determine

similarity. Therefore protein products which are manufactured by the independent

manufactures would never be identical to the innovator product, but at maximum would be

similar to innovator molecule possessing the same clinical attributes despite of not being the

same molecule (Kresse, 2009).

An ideal legal and regulatory process which allows the approval of the biosimilar

products has to attain equilibrium between various factors which involve the facility for market

entry for biosimilars, competition for products which have lost patent protection, to promote

research and development and provide incentives, and finally the most important is to evade

any risk associated with patient safety. Though it is considered that advent of biosimilars will

lead to reduction of redundant or even unethical clinical trials of animals as well as human and

biosimilars will propose economic benefits but biosimilars do not bring any innovative medical

progress as the original product is already available proving effective and safe to the patients.

Therefore the approval pathway for biosimilars should ensure that appropriate standards of

same level as innovative products should be maintained. Usually it is agreed that the regulatory

pathway for traditional low molecular generic drugs is not suitable for biologic or

biopharmaceutical medicines. There are initiatives and regulatory pathways already established

or under development in various regions of the world (Kresse, 2009).

Biosimilars are licensed and marketed in various regions of the world including the less

regulated markets such as India, China and South Korea. The category of the products which are

marketed in these regions include interferons, EPOs, growth hormones, enzymes, interleukins,

monoclonal antibodies etc. As compared to these less regulated markets there is a significantly

less number of biosimilar products available on the European market with only few categories

Page 28: BIOSIMILARS: SCIENCE TO MARKET

19

of products such EPO, filgrastim and somatropin. On the other side, however, there is the

establishment of a highly developed framework of regulations in the European Union for the

approval of biosimilar products. The regulatory framework of Europe by the EMA (European

Medicines Agency) (earlier called as EMEA, terms used alternatively in this report) has

influenced the regulatory authorities in other countries like Unites States, Japan and Canada

which will work on the similar lines (Zuniga & Calvo, 2009).

3.1 REGULATORY FRAMEWORK EUROPE:

The regulatory framework and process for the biosimilar product approval is defined by

the European Directive 2001/83/EC which is modified by Directive 2003/63/EC and Directive

2004/27/EC. These Directives provide detailed specific guidelines which have to be followed by

the biosimilar developers. These guidelines include an overarching guideline (Box 1) and also

other broad guidelines which are associated with quality of the product and clinical and non

clinical data which is to be provided by the biosimilar applicant (Fig 3.1). EMEA has also provided

product specific guidelines and are also in the course of developing supplementary guidelines

which will upgrade the main guidelines as new information is continuously added and found.

Box 2 states the guidelines which are being issued in the European Union in reference to

biosimilars.

Fig 3.1: Regulatory guidelines. Source: Kox, S., 2009

Page 29: BIOSIMILARS: SCIENCE TO MARKET

20

Box 1: Summary of biosimilar overarching guideline (EMEA/CHMP/437/04)

Source: Taylor, P., 2009.

Page 30: BIOSIMILARS: SCIENCE TO MARKET

21

Box 2: Summary of biosimilar overarching guideline (EMEA/CHMP/437/04)

PD= Publication date; ED= Effective Date. Source: Taylor, P., 2009.

Page 31: BIOSIMILARS: SCIENCE TO MARKET

22

As per the legislation the Committee for Medicinal Products for Human Use (CHMP) of

the European Medicines Agency (EMEA) has discretion to develop guidelines which determine

the amount of clinical trials required as per the product (Chu & Pugatch, 2009).

The pharmaceutical products resulting from biotechnology are registered in Europe

alone through a centralized procedure which results in a European Union license. The European

Union license is valid in all the member countries of EU (Zuniga & Calvo, 2009).

The biosimilar developers for the marketing authorization of the product have to fulfill the

following requirements which are set by EMEA:

Data on the comparability studies should be provided between the applicant biosimilar

product and reference innovative medical product.

Non clinical studies data which are generally required in limited details as compared to

applications for the innovative product.

To prove safety and efficacy of the biosimilar product clinical studies are required.

An obligation to provide post market pharmacovigilance arrangements as part of the

approval process (Zuniga & Calvo, 2009).

EMEA has developed biosimilar regulatory guidelines with major distinctions from the

generic medicines approval process considering the fact that biosimilars are not expected to be

identical to the innovator biologic medicines. The fact that conventional generics are different

from biosimilars is recognized by European legislation and mentioned in the Article 10(4) of EU

Directive 2001/83/EC which has been modified by Directive 2004/27/EC (Kresse, 2009).

Establishing bioequivalence alone of the biosimilars to the innovator biologic is not sufficient

to get market approval. The basic fundamental idea behind the guidelines is that the biological

activity of the biosimilars cannot be determined if the active substance is different from the

innovative biologic. Analyzing the pharmacokinetic properties of the biosimilars will not offer

satisfying results on whether the similar but not identical nature of the active substances has

lead to changes in clinical safety and efficacy profile. Therefore the biosimilar applicant to

demonstrate the similar product quality, safety and efficacy profile to the innovator biologic has

Page 32: BIOSIMILARS: SCIENCE TO MARKET

23

to perform further clinical and non clinical studies according to the EMEA clinical and non

clinical guidelines (Box 3) to acquire market approval (Fox, 2010).

Box 3: Summary of Biosimilars Clinical & non clinical Guideline Principles for MEA/CHMP/42832/05

Source: Zuniga & Calvo, 2009

Page 33: BIOSIMILARS: SCIENCE TO MARKET

24

3.1.1 PRODUCT SPECIFIC GUIDELINES:

Apart from general regulatory guidance issued for all biosimilar categories, EMEA has

additionally issued guidelines which are specific to the product class such as EPO, G-CSF, human

soluble insulin, somatropins, low molecular weight heparin and interferon alpha. The guidelines

for other product category such as monoclonal antibodies or biosimilar medicinal products

containing monoclonal bodies have been in the stages of finalization. The Biosimilars Medicinal

Products Working Party (BMWP) is in the process of preparation of the guidelines for the Similar

Biological Medicinal Products containing Follitropin alpha and beta-Interferon. Currently

according to the EMA (2010) there is also revision and maintenance of the existing overarching,

non-clinical and Clinical guidelines by Biosimilars Medicinal Products Working Party (BMWP).

The product specific guidelines explain in details the extent and type of studies, both in

terms of clinical and non clinical, which should be carried out and presented to the regulators to

obtain approval for the product. The guidelines not only expect the biosimilar product to be safe

and effective but also to be comparative in nature and most importantly to identify variation in

response between the biosimilar and innovative biologic. It is of important concern that the

minute differences of biosimilars to innovative product with reference to quality are not

acceptable by the guidelines and even if they are anticipated then proper explanation regarding

the implications caused by the variation should be provided. As per the guidelines, it is crucial

that the biosimilar applicant demonstrates that the quality, safety and efficacy of the biosimilar

are similar to that of the innovators biologic it seeks to copy. The attributes of the biosimilars

should display equivalence and cannot be worse, better or different from the innovator biologic

product. If the biosimilar is different or better than the original biologic, which is not an

alternative as it implies there is lack of similarity, then the biosimilar product may have to be

withdrawn from the application process, would get rejected or have to apply for new product

application necessitating a need to pursue a full stand alone pathway (Fox, 2010).

Given that the data submitted by the biosimilar applicant are less than the innovator,

the approach of the EMEA to acquiring authorization of a biosimilar, in that, there is no

standard data set which is applicable to all classes of biologics, is sensible. There is a variation

Page 34: BIOSIMILARS: SCIENCE TO MARKET

25

among different classes of biologics in terms of benefit and risk profile, whether surrogate

markers are available and validated, adverse events possible and clinical indications. EMEA

provides product class specific guidelines that suggest the data and studies which are supposed

to be conducted but does not provide the exact equivalence margins. Therefore the guidelines

do not lay down set standards for approval and there is possibility to maneuver when it comes

to settling exact standards for approval of any biosimilar (Fox, 2010).

The Regulatory pathway for biosimilars in the European Union was in effect from 2005

and since then there have been 14 biosimilar approvals and 7 products received negative

opinion or were rejected by the EMEA (Fig 3.1 & 3.2). The biosimilar applicant requires clinical

studies consisting of 200 to 500 subjects which depend on the product class of the biosimilar. In

contrast the innovator biologics have to conduct clinical trials on thousands of subjects to

achieve a range of clinical indications. This indicates the extent of reduction of clinical trials for

the biosimilar applicant (Fox, 2010).

Fig 3.2: market exclusivity. Adapted from Kox, S., 2009

Page 35: BIOSIMILARS: SCIENCE TO MARKET

26

3.1.2 IMMUNOGENICITY:

The generic drugs are different from the biosimilars in various aspects and one critical

aspect is their capability to produce immune response. A change in immunogenicity profile is of

most important concern as it can have enormous consequence on the product safety (Zuniga &

Calvo, 2010). The implications which are caused by immunogenicity are difficult to predict. The

formation of antibodies can either have harmless clinical effects or can result into serious

diseases and some cases considerable adverse events. Immunogenicity effects can be explained

by the example of Eprex which is an EPO product marketed by Johnson & Johnson in the

European Union with no significant immunogenic concerns for almost 10 years prior to 1998

when regulatory bodies requested for change in the product. Johnson & Johnson modified the

Eprex formulation by interchanging human serum albumin with polysorbate 80 and glycine

which resulted in pure red-cell aplasia (PRCA). Pure red-cell aplasia is a severe type of anemia.

The antibodies which are produced due to Eprex neutralize all the exogenous rHuEPO and also

cross react with endogenous erythropoietic proteins which results into ineffectiveness

erythropoiesis and serum EPO is not detectable. J&J later found out that the polysorbate 80 in

the single use syringes reacted with rubber stoppers to leach plasticizers which triggered the

immune reaction and caused PRCA. In 2003 there was 90% reduction in PRCA by changing

uncoated rubber stoppers to Teflon coated rubber stoppers (Chen, 2009).

The EMA approval process in association to immunogenicity concerns with biosimilar

products can be explained with clinical and non clinical testing and comparability decision of the

biosimilar product Retacrit® with the innovator product Eprex®/Erypo®. The applicant offered a

database of studies which was considered as sufficient by the EMA which consisted of a report

on studies of clinical immunogenicity within a time period of twelve month taking data from 227

subjects with renal anemia which was later updated to additional 585 subjects. The toxicity,

pharmacokinetic and pharmacodynamic analysis and biologics safety and efficacy are often

affected due to anti-drug antibody (ADA) reactions. The safety issues are also related with

neutralizing ADA (nADA). In case of Retacrit® and reference product Eprex® the serum samples

were acquired for the determination of ADA before dosing and through the safety studies. A

Page 36: BIOSIMILARS: SCIENCE TO MARKET

27

validated radioimmunoprecipitation assay was used for the testing of the Anti-EPO antibodies

which indicated that there was a low occurrence of ADA in the subjects which were treated

either with the biosimilar Retacrit® or the innovator product and also no patients who were

ADA positive showed signs of PRCA. These results indicated that there was no need for the

NADA tests, but for post marketing surveillance a validated nADA testing was available. The

EMA has not concluded on the specific value of the predictive immunology but its usage is

recommended. For example the transgenic mouse models could be helpful to estimate

probable immunogenicity of the protein in question. Generally in preclinical immunogenicity it

is difficult to recognize that the estimations are relevant without clinical data to validate

preclinical assessments (Barbosa, 2011).

The European biosimilar regulatory pathway has specific consideration towards the

biosimilars immunogenicity issues and post marketing activity to identify potential concerns.

The estimation of immunogenicity of biosimilars cannot be determined by only preclinical trials.

As a result clinical trials along with a post marketing surveillance plan are mandatory for the

authorization of biosimilars (Zuniga & Calvo, 2009). In context to the persistent chronic

treatment, there is a requirement of immunogenicity data for about a year of treatment before

authorization (EMEA/CHMP/BMWP/42832/05).

3.1.3 EXTRAPOLATION:

With the appropriate justification, as per the relevant guidelines, the extrapolation of

clinical data, for indications for which the drug has not been evaluated in clinical trials, has been

allowed for biosimilars. According to guidelines provided by the EU, extrapolation is not

allowed, but is considered on a case by case basis based on various factors such as complexity of

the product and mechanism of action etc. CHMP guidelines allow extrapolation based on

known mechanism of action and the sensitive indication where, if significant differences would

exist between biosimilar and the reference product, they would be detected in that particular

population (Ruiz & Calvo, 2010). The extrapolation of safety is not approved for any indications.

EMEA has approved recombinant granulocyte –colony stimulating factor for the reduction in

Page 37: BIOSIMILARS: SCIENCE TO MARKET

28

neutropenia after cancer chemotherapy, but the approval of other indications of reference

product were made by extrapolation considering that mechanism of action of biosimilar is the

same. (Zuniga & Calvo, 2010).

3.2 EU BIOSIMILAR APPROVAL PROCESS:

According to the European Directive 2004/27/EC the conducting of the comparability

studies between the biosimilar and the reference product is necessary but the requirements for

probable test are not addressed. The comparability exercise is of various types which include

physiochemical, biological, Pre-clinical and clinical. The reference biologic product under

consideration must be a medical licensed product on the basis of the complete document as per

the necessities of article 8 of Directive 2001/83/EC modified by Directive 2001/83/EC (Zuniga &

Calvo, 2010). The reference product chosen to compare with the applicant should be same

throughout the comparability programme. Comparability should be in terms of product quality

and manufacturing process as the safety and efficacy of the product is directly associated to the

manufacturing process.

3.2.1 COMMON TECHNICAL DOCUMENT:

The biosimilar applications should be made completely in agreement with the Common

Technical Document (CTD) presentation (CPMP/ICH/2887/99). It is structured into five different

modules which biosimilar applicants have to follow as shown in Table 3.1. The information to

be provided is not restricted to first 3 modules, but additional data will be required. Generally

the supplemental data is determined on a case by case basis in relation to specific scientific

guidelines (Directive 2003/63/EC) (Zuniga & Calvo, 2010).

Page 38: BIOSIMILARS: SCIENCE TO MARKET

29

Table 3.1. Format of the dossier- modules of the CTD. Source: Zuniga & Calvo, 2009

3.2.1.1 MODULE 1:

In module 1 a brief document is to be submitted comprising information about the

details of the product, the manufacturing process involved, raw materials used, and its active

substance. It also involves other information about the comparability exercise such as changes

made during development which would affect the safety and efficacy and detailed description

of the reference product.

3.2.1.2 MODULE 2:

Module 2 expects the data on normal requirements which includes the general idea and

synopsis of the quality, clinical and non-clinical data.

3.2.1.3 MODULE 3:

A complete quality document which provides information on chemical, pharmaceutical

and biologic information is required for biosimilars. In addition to this information,

comparability studies should be provided as per the guidelines of EMEA.

Page 39: BIOSIMILARS: SCIENCE TO MARKET

30

3.2.1.4 MODULE 4:

The non-clinical studies to determine the differences and similarities between the

applicant product and reference product are to be demonstrated in module 4. It is crucial to

identify the biological product characteristics of the biosimilar based on studies related to

physiochemical and biological characterization.

3.2.1.5 MODULE 5:

To demonstrate the safety and efficacy of the biosimilar at the clinical level the studies

conducted at non clinical level are not sufficient. It is essential to submit the design of the

clinical program to the regulatory agency. The extent of the biosimilar trial depends on the

specific class of the product (Zuniga & Calvo, 2010).

The approval of the biosimilars in the European Union will lead into formation and

publication of public report which is called European Public Assessment Report (EPAR). The

EPAR is designed for the public and is written in collaboration with the biosimilar applicant. The

main purpose of the report is to explain and provide the transparency of the biosimilar

application and regulatory process involved during the approval period (Zuniga & Calvo, 2009).

3.3 UNITED STATES REGULATORY FRAMEWORK:

The Regulations framework of Biologics in the United States is regulated by Public Health

Service Act (PHSA), but for few exceptions such as insulin and human growth hormone which

come under the Food Drug and Cosmetics Act (FDCA). FDCA has a regulatory pathway for the

generic drugs of the conventional chemical drugs, but PHSA does not have any approval system

for the generic versions of biologics (Clark, 2009). According to the standards of FDCA, the

biological drugs are authorized on the basis of identity, effectiveness and purity rather than on

efficacy and safety. Even though there are no biosimilar approval options by the PHSA, there are

still few biosimilar products out in the United States market. For example Omnitrope by Sandoz

a biosimilar version of recombinant human growth hormone which is similar to Genotropin by

Pfizer was authorized by the FDA in 2006 for the United States. The approval of Omnitrope was

Page 40: BIOSIMILARS: SCIENCE TO MARKET

31

made through a New Drug Application (NDA) which used the route 505(b) (2). This route is

different from the generic approach which is called as Abbreviated New Drug Application

(ANDA) in various ways such as there is no need for sameness which gives room for satisfactory

similarity. The applicant using the 505(b) (2) route can use the available research which implies

that the FDA without citing the trade secrets of Pfizer could evaluate Sandoz’s Omnitrope (Clark,

2009). The complicated biosimilar products such as interferons cannot be approved through this

regulatory pathway.

President Obama approved the “Patient Protection and Affordable Healthcare Act” in

March 2010. The main objective of this act was to form legislation by designing a regulatory

pathway which will save healthcare cost and creating a flexible route for the approval certain

biologic which reduces the cost of development. The 351 (k) route is the new regulatory

pathway for biosimilars which is provided by the Biologics Price Competition and Innovation Act

(BPCIA) as part of the Affordable Care Act. According to the pathway the biosimilar should be

compared to the single innovative reference product which is authorized under 351 (a) route.

Two types of products will be provided by this pathway, Biosimilar and Interchangeable

biosimilar. For obtaining the interchangeable biosimilar approval exact guidelines and

requirements are under discussion. The manufacturers should also comply with patent

disclosure arrangements as per the act. The authority of describing the guidelines for regulatory

framework is given to FDA and they have not yet revealed data requirement for approval

(Greer, 2011).

3.4 GLOBAL LANDSCAPE:

International Conference on Harmonization (ICH) aims to synchronize the approval

process and regulatory requirements of drug or biologics in the United States, EU and Japan.

Biosimilar regulatory framework is already established in EU and Japan but the legislation is still

under discussion in US (Chen, 2009).

There is establishment of the regulatory pathway have taken place in various countries

of the world such as Brazil, Taiwan, Mexico, Argentina, India, Canada and South Africa. There is

Page 41: BIOSIMILARS: SCIENCE TO MARKET

32

a direct acceptance of the guidelines of EMEA in Australia and on similar basis regulation are

established in Malaysia, Japan and Turkey. The approval pathways followed by various

countries are not clear in terms of scientific reasoning, therefore the World Health Organization

(WHO) adopted a guideline to evaluate similar biologic products which will result in availability

of the regulated biosimilar products worldwide (Kresse, 2009).

Page 42: BIOSIMILARS: SCIENCE TO MARKET

33

4. CASE STUDY-BIOSIMILAR INSULIN:

In general biologics are complex molecules to produce and biosimilar insulins present

special challenges. Their therapeutic window is narrow and the accuracy of their dosing is

dependent on the product formulation and quality of the administrative device. Therefore for

these specific reasons EMA has issued strict guidelines which the biosimilar applicant must

follow to receive approval of the biosimilar soluble insulin (Sauer, T. and Kramer, I., 2010).

4.1 MANUFACTURING:

The recombinant human insulin production is a highly complicated process (Fig 4.1). The

first step is the isolation of the human insulin gene which has specific sequence which codes for

the human insulin. After the isolation the gene is attached to the vector and then it is inserted

into a host cell which is generally E.coli or a yeast species. The recombinant cells which are

formed are screened which results in the formation of the master cell bank, then further

cultured and fermented. After fermentation the protein which is produced is isolated, purified

and is folded in order to form secondary structure. To achieve biologically active insulin, the

secondary structure is enzymatically cleaved. Different adsorption and chromatographic

techniques are employed for the purification of the recombinant insulin. To prevent the insulin

from losing its biological activity or avoid aggregation of the product and bacterial growth, the

purified product is subjected to crystallization, lyophilization and formulated by addition of

other compounds such as protamine is added for long acting formulation (Marre & Kuhlmann,

2010). If there is any change in the various sequential process of insulin production such as

change in vector selection or change in formulation will result in an insulin product which will be

identical to the innovator insulin product in terms of structure and amino acid sequence but its

clinical properties will differ from the innovator product.

Page 43: BIOSIMILARS: SCIENCE TO MARKET

34

Fig 4.1: Post fermentation steps in manufacturing process. Source: (Marre, M. & Kuhlmann, M., 2010)

4.2 EMEA REQUIREMENTS:

For the market approval of the soluble insulin biosimilars, EMEA has laid down specific

guidelines which explain the requirements to be fulfilled by the applicant. As per the guidelines

like all the biosimilars, insulin as a biosimilar product should be analyzed with the comparative

technique to prove equivalence with the reference product. For biosimilar insulin approval the

preclinical studies are required which consist of in vitro pharmacodynamic studies, in vitro

affinity bioassays and receptor binding assay for insulin as well as IGF-1.

Page 44: BIOSIMILARS: SCIENCE TO MARKET

35

The requirement of EMEA is at least one pharmacokinetic single dose crossover study in

patients suffering from type 1 diabetes by subcutaneous administration to compare the

biosimilar with the reference product. To check the biosimilar insulins hypoglycaemic response

profile, clinical activity must be determined in pharmacodynamic study designed as a double

blind crossover, hyperinsulinaemic, euglycaemic clamp study (Sauer and Kramer, 2010). For

biosimilar insulin clinical efficacy trial is not needed but there is a requirement of clinical safety

study. For at least the period of 12 months, the insulin product immunogenicity should be

inspected through the clinical studies. Six months of comparative phase should be included in

the clinical trials. To detect any clinically important immunogenicity that may occur in the long

term, the developers should design a pharmacovigilance plan (Marre & Kuhlmann, 2010).

4.3 MARVEL’S INSULIN REJECTION:

Marvel Life Sciences Ltd in March 2007 submitted a biosimilars application for market

approval of recombinant insulin in three different formulations. Marvel Life Sciences Ltd

presented their data from their studies intended to show the similarity between Marvel’s insulin

and the reference insulin product in experimental models and in humans. The consequence of

Marvel’s insulin on the blood sugar levels was studied in Twenty-four healthy volunteers with

that of the reference insulin product and these studies were presented to the EMEA. Another

important study was also presented which involved 526 diabetes mellitus patients who either

received Marvel’s insulin or reference insulin for the period of 12 months.

There were various issues found by CHMP regarding the data and application submitted

by Marvel Life Sciences. CHMP found that data on many critical aspects of the application were

not enough and unclear. Review of the application was done by EMEA, considering the

application CHMP formed a conclusion that the three formulations of biosimilar insulins by

Marvel Life Sciences Ltd were not comparable with the reference insulin. The CHMP rejected

Marvel’s insulin on the grounds of Quality, Clinical and Non-Clinical aspects.

Page 45: BIOSIMILARS: SCIENCE TO MARKET

36

4.3.1 QUALITY ASPECTS:

The Proper evaluation of the application was not possible because sufficient amount of

data was not submitted on the development and manufacture of the drug substance as well as

the drug product. There was confusion whether the reference product used for the

comparability exercise was valid or not. The explanation provided for the process like

fermentation, harvesting, purification and modification in the application were not in complete

details. The comparability exercises to detect impurities in the insulin product to that of the

reference product were not adequate to draw a conclusion that Marvel’s insulin was biosimilar

to the reference insulin. There was a huge confusion that the dossier submitted during

application was unable to specify two different presentations of the drug product (vials and

cartridges).The important details such as drug substance batch number, size and site of

manufacture and details of where the batches have been used for clinical and pre-clinical trials

were absent in the dossier (Joshi, 2009).

4.3.2 NON CLINICAL ASPECTS:

Based on the data submitted, the committee was not able to review the comparability of

Marvel’s insulin with the reference product due to insufficient explanation.

4.3.3 CLINICAL ASPECTS:

The pharmacodynamic studies did not provide the result of lowering the blood glucose

level as compared to the reference product. The sufficient pharmacokinetic studies were not

carried out such as single dose crossover comparative studies by the use of subcutaneous

injection as per the guidelines. The immunogenicity of the insulin product was not completely

evaluated and validated. Additionally the pharmacovigilance plan presented in the dossier was

not up to the requirement of the EMEA guidelines (Joshi, 2009).

Due to inadequate studies and lack of well presented data in January 2008 it was

declared by the EMEA that Marvel Life Sciences Ltd have withdrawn applications for all three

insulin formulations (Marre & Kuhlmann, 2010).

Page 46: BIOSIMILARS: SCIENCE TO MARKET

37

5. MARKET ANALYSIS:

The focus of the market analysis in this thesis is based on major markets such as the

United States and the European Union.

5.1 BIOSIMILARS ON MARKET:

The expiration of patents of a number of first generation of biologics has led to the

approval of various biosimilar drugs by the European Medicines Agency (then EMEA, now EMA)

in Europe. EU in 2003 held discussions on the follow on biologic recombinant proteins concept

and then later the guidelines on biosimilars were established and took effect in 2005. The

Committee for Medicinal Products for Human Use (CHMP) according to the guidelines for

biosimilars requires complete characterization in terms of physical, chemical and biological of

the biosimilar product as compared to the reference product. To prove the safety and efficacy

of the biosimilar product widespread characterization, clinical and non clinical data is required

but as per the guidelines the amount of data required will be less than the application of the

innovators drug (Greer, 2011). Omnitrope, a biosimilar version of somatropin was the first

biosimilar drug to get approval from the EMEA in April 2006. Another Human growth hormone

called Valtropin was approved by EMA immediately two weeks after the approval of Omnitrope.

Table 5.1 illustrates that to date the EMA has approved 14 biosimilars products which include

the versions of somatropin, EPO and filgrastim. Nine different biosimilar companies have

successfully launched 7 biosimilar molecules under 14 different trade names (EBE 2010).

Page 47: BIOSIMILARS: SCIENCE TO MARKET

38

Table 5.1: Biosimilars approved by the EU. Source: Greer, F.M., 2011

Table 5.2 shows that several applications such as interferon alpha-2a and insulin

received negative opinion and some applications were not successful, either rejected or

withdrawn voluntarily.

Page 48: BIOSIMILARS: SCIENCE TO MARKET

39

Table 5.2: Unsuccessful biosimilar applications in the EU. Source: Greer, F.M., 2011

In the United States there is no legislation for a clear regulatory approval pathway for

biosimilars but still Omnitrope, a biosimilar version of somatropin was authorized by the use of

the Abbreviated New Drug Application (ANDA) process under the Hatch-Waxman Act following

EU approval (Horikawa et.al, 2009). There were various biosimilars approvals before Omnitrope

which didn’t achieve to receive as much attention as Omnitrope. These approvals include

different recombinant biologic drugs with trade names such as Glucagen, Hylenex, Hydase and

Amphadase. Fortical by Unigene which is similar to Miacalcin by Novartis used for the treatment

of osteoporosis was the first biosimilar recombinant DNA product to be authorized by FDA by

the 505(b) (2) route under NDA (Clark, 2009).

Page 49: BIOSIMILARS: SCIENCE TO MARKET

40

5.2 MARKET SIZE & GROWTH:

The patent expirations from the year 2009 through 2013 are expected to trigger the

battle the approval and production of biosimilars (Crandall, 2009). The potential of the total

worldwide market for the biosimilar products is quite significant amounting up to several billion

dollars annually. Significant opportunity for the biosimilars market is also created as the

biologics worth $25 billion are expected to go off-patent by 2016 (Business Insights Ltd, 2009).

The biologic market has outperformed the pharmaceutical market which is driven by high prices

for the therapies which cannot be managed by traditional drugs. In 2007 Biologics contributed

more than 10% of the global pharmaceutical revenues. The annual rate of growth for the

biologics is growing at the rate of 12%-13% which is almost double the global pharmaceutical

industry rate of growth (Business Insights Ltd, 2009).

The growth of the biosimilars market is also fueled by the rapid penetration of the novel

biologics in the global pharmaceutical markets and the gradual expiry of the patents of the

novel biologics. There was 5.9% growth in the global biosimilars market in 2007 to reach the

value of approximately $1 billion (Business Insights Ltd, 2009). The growth of the biosimilars will

be majorly driven by the four drug classes - erythropoietin (EPO), filgrastim, human Growth

Hormone (hGH) and insulin in the future. The revenues from the biosimilars are currently less

because one of the profitable markets, such as the U.S. is facing regulatory restrictions but after

the regulatory framework is established there will be a number of products which could have

market authorization and thereby increasing the revenues and size of the biosimilar market

(Crandall, 2009).

5.3 MARKET POTENTIAL:

There is a range of reports which provide estimates on worldwide biosimilars market

figures and forecast. As this market is highly speculative the range of figures and estimates

provided by the various reports vary.

Page 50: BIOSIMILARS: SCIENCE TO MARKET

41

The total global biosimilar market potential for the period of 2006-2013 is forecasted for

the currently expired patents by Crandall (2009) which indicates that by the year 2013 the

revenues generated would be $358 million at the 17.0 percent growth rate and for the period of

2006-2013 the compound annual growth rate would be 32.5% (Table 5.3).

Table 5.3: Total World Biosimilar Market Potential 2006-2013 (Products with currently expired patents).Source : Crandall M., 2009.

Fig 5.1 represents the steady growth of the global biosimilar market and its potential

with currently expired patents.

Page 51: BIOSIMILARS: SCIENCE TO MARKET

42

Fig 5.1: World Biosimilar Market potenatial by region 2006-2013, products with currently expired patents. Source: Crandall, 2009.

5.4 REGIONAL MARKET ANALYSIS:

The European Union and the U.S. are the major markets for the sales and revenue

generation from the biosimilars. As compared to any other region worldwide the products in

the U.S. perform better in reference to sales and the drug approval in the U.S. generally sets the

standard for the approval abroad. Marketers consider that the U.S. market is generally most

favorable. Lack of regulation is holding back the biosimilar market from expanding in the U.S.

Although in the U.S. there is no clear developed pathway for the authorization and approval of

the biosimilars as compared to other countries, currently only few approvals of biosimilars can

be seen for the regions other than the U.S. There is some progress with a range of biosimilars

approvals in markets such as Eastern Europe, Asia and South America but still the sales revenues

are quite less as compared to the major markets such as the U.S. and Europe. Novartis, is the

main competitor in the biosimilar market, as this company has made progress in early phase of

Page 52: BIOSIMILARS: SCIENCE TO MARKET

43

the market. In less regulated and less developed markets like India, Rituxan and Neupogen are

substituted by the generic counterparts (Crandall, 2009). Table 5.4 shows the distribution of

market potential for biosimilars in major markets such as Europe and the United states and the

rest of the world from currently expired patents.

Table 5.4: World Biosimilar Market Potential by Region 2006-2013 (Products with currently expired patents). Source: Crandall M., 2009.

5.5 BIOLOGIC CLASS MARKET ANALYSIS:

Table 5.6 shows the forecast for market potential and revenue generated from individual

product categories which are currently marketed. The biosimilar versions of the blood products

like erythropoietin and G-CSF are in great demand in the global biosimilars market. Sales

revenues generated from these products alone in the period of 2008 were estimated to be $62

million (Crandall, 2009). This sales figure consists of the sales of the products in the European

Union as well as in the less regulated market throughout the world where relaxed laws of

Page 53: BIOSIMILARS: SCIENCE TO MARKET

44

biosimilars exist. Despite of the fact that insulin is the favorable for biosimilar production due to

the reasonably less complex manufacturing process, the sales of the biosimilar insulin is

relatively low as compared to the total market of insulin. Another key product in biosimilar

products is HGH which is favorable for production in the U.S. and Europe, but the sales results

are much lower than predicted by producers. Global sales for the biosimilar HGH is about

$15million for the year 2008 and with the growth rate of 21.9% during the projected period

(Crandall, 2009). The category of biosimilar drugs which involves autoimmune and oncology

products shows a rapid growth with sales estimated to be $39 million throughout 2008

(Crandall, 2009). The accessibility of the monoclonal antibodies and multiple sclerosis

therapeutic biosimilar versions is found in regions of the world where there is less regulation

and the patent laws are not strict.

Table 5.6: The world market potential for Biosimilars by Biological Class (EPO, G-CSF, insulin, Interfereon, alpha, others) 2006-2013. This forecast includes currently marketed classes only. Source: Crandall M., 2009.

Page 54: BIOSIMILARS: SCIENCE TO MARKET

45

It has been reported by (Emmerich, 2010) that by the year 2015 the European and

United States biosimilar market size could reach US$ 10 billion. The monoclonal antibody (mAB)

segment is anticipated to generate most revenues. The Biosimilars’ largest market share is

expected from the revenues generated from mAB such as Remicade and Rituxan. Fig 5.2

indicates the predicted market share among various classes of biosimilar drugs by 2015. The

market is dominated by various companies which will lead in variation in market penetration

between products. For example biosimilar insulin market penetration is considered low

because the market is dominated by other three originator companies and on top of that

advanced injection system is required for insulin.

Fig 5.2: Expected Biosimilar market split in 2015 Source: Emmerich, R. (2010)

5.6 MARKET OPPORTUNITIES:

The global biosimilar sales estimates (Fig 5.3) were reported by Clark (2009) for a period

of five years ending in 2012. The estimates provided in the report were based on biosimilar

activities in Europe as the US market, due to lack of regulatory framework, is not likely to show

any momentum during this period. Even though the biosimilar market value would be around

$13 billion for the period 2009-2012 which is quite considerable, this biosimilar market is

Page 55: BIOSIMILARS: SCIENCE TO MARKET

46

actually predicted to represent only a small percentage of total pharmaceutical and generic

sales of the future.

Fig 5.3: Forecast of the global biosimilar market value in $billion: 2008-12. Source: Clark, T.D., 2009

The prices of the biosimilar products will generally be 20% to 30% less than the

corresponding innovator products. The average price of biologic could be $16,425 p.a. which is

around 20 times the cost of the chemical generics (Emmerich, 2010). As compared to the 90%

savings from traditional generics the savings of 30% from biosimilars is not significant, but, if

considering a situation where the treatment of the metastatic cancer through the biologic drug

can cost up to $200,000 a year, savings of mere 30% amounts to much more than a 90% savings

on a drug which costs $1000 (Emmerich, 2010). The biologic drugs are expensive at an average

daily cost of $45 or 22 times that of conventional drugs. Table 5.7 provides the estimates of the

treatment cost per patient of selected biopharmaceuticals. The first wave of the

biopharmaceutical drugs accounting up to $10 billion market have already lost patent

protection and by 2018 further biopharmaceutical drugs of $20 billion market will lose patent

protection (Clark, 2009). These figures and circumstances has led to immense interest towards

the market opportunities generated by the biosimilars industry.

Page 56: BIOSIMILARS: SCIENCE TO MARKET

47

Table 5.7: Estimates of treatment cost per patient of selected biopharmaceuticals. Source: Crandall, 2009.

5.6.1 PATENT EXPIRY:

The expiry or pending expiry of patents of the biopharmaceuticals products such as

interferons, human growth hormone and epoietins is the most serious problem faced by the

biopharmaceutical industry. The expiry of patents of many blockbuster biologic products has

created immense market opportunities for biosimilar industry in markets where the innovator

companies are already established. When it comes to patent protection, there are numerous

patents which are generally issued by the innovator companies for specific APIs (Active

Pharmaceutical Ingredient The extent to which the innovator companies can go to protect and

extend patent protection of these patents poses extreme difficult issues for the prospective

biosimilar companies (Taylor, 2009).

In the United States the Congressional Budget Office has estimated that out of the $40

billion exhausted on the biopharmaceutical products in 2007, the products which contributed to

Page 57: BIOSIMILARS: SCIENCE TO MARKET

48

the three quarters of the spending will lose patent protection by the year 2019. The government

initiatives will be benefited by the cost reductions through the period of 2010-2019 which

would amount up to $9.1 to $11.7 billion and during this period the private insurance programs

would experience 0.2% reduction in premiums (Clark, 2009).

Reports have mentioned that there are major opportunities for the biosimilar

manufacturers during the period of 2010 to 2015 as throughout this period 45 biologic drugs

patent will expire and their value is more than $60 billion in global sales (Emmerich, 2010). Fig

5.4 shows the number of biologic drugs set to lose patent protection per year during the period

of 2010 to 2015 and annual global sales.

Fig 5.4: Number and value of biological drugs set to lose patent protection per year through 2015 Source: Emmerich, R. (2010)

Page 58: BIOSIMILARS: SCIENCE TO MARKET

49

Table 5.8 shows that the blockbuster biologic drugs such as Enbrel, Remicade, Rituxan

with the global sales in billions lose the patent protection in major markets like US and Europe

from 2012 to 2015.

Table 5.8: Blockbuster biological drugs set to lose patent protection per year through 2015. Source: Emmerich, R. (2010)

5.6.1.1 PATENT EXPIRY BY BIOPHARMACEUTICAL CLASS:

5.6.1.1.1 ERYTHROPOIETINS (EPOS):

The biologic drugs have different classes based on the therapy areas. In the

erythropoietins (EPOs) category there are many products which have been repeatedly reported

to have gone off patent in December 2004. First generation EPOs by Amgen such as Epogen

(epoetin alfa) and Johnson & Johnson’s Procit and Eprex have gone off patent. It has been

reported that Amgen is also involved in patent disputes on various development patents of EPO

with Wyeth, Roche. Various disputes are resolved but the terms and agreements are not

disclosed which could indicate that parties resolved disputes by cross licensing their patents

(Taylor, 2009).

5.6.1.1.2 INTERFERONS ALPHA (2A & 2B):

Interferons Alpha market segment has two fundamental products Pegasys and

PegIntron. The usages of these two products are done in the treatment of hepatitis C

frequently with the combination of ribavirin. Standard interferon products such as Roferon

(interferon-ά 2a) and Intron-A (interferon-ά 2b) which were introduced before Pegasys and

Page 59: BIOSIMILARS: SCIENCE TO MARKET

50

PegIntron are still marketed but are not as effective as PEGylated products (Pegasys and

PegIntron) and so they are prescribed to a lesser extent than PEGylated products(Taylor, 2009).

Table 5.9 shows the expiry dates of the patents of major Interferon Alpha products

which currently exist on the market and indicates use of improved method such as pegylation as

a strategy to lengthen the market exclusivity following the expiry of patents of the standard

interferon products as both the standard and pegylated interferon products are from same

companies.

Table 5.9: Interferons on market and patent expiries. Source: Taylor, P. (2009)

5.6.1.1.3 INTERFERONS BETA (1A & 1B):

Some most important patents have recently expired of a class of biopharmaceuticals

which constitute of interferons (beta-1a and beta-1b). For the treatment of Relapsing/Remitting

Multiple Sclerosis the principal product used is Avonex which belongs to key group of

interferons and is manufactured by Biogen Idec. For the production of beta interferon there are

various companies which have pending patent applications or issued patents in the United

States, Europe and other major market and countries and these patents are regarded as the

Taniguchi patents. There also some other patents which are called as Roche patents and the

Rentschler patents which are pending patent applications or issued patents for interferon beta

by the companies EMD Serono Pfizer and Bayer. Biogen Idec has access to rights in different

countries and markets of the world such as United States, Europe and Japan for production and

marketing of Avonex as per the Taniguchi, Roche and Rentschler issued patents (Taylor, 2009).

Page 60: BIOSIMILARS: SCIENCE TO MARKET

51

The Taniguchi patents are going off patent in the United States in 2013 and they have

already expired in other parts of the world. The Roche patents have expired in most countries of

the world and they will expire in May 2008 in the United States. The European Union Rentschler

patents expire in July 2012. Other interferon beta-1a products such as Betaferon (Betaseron) by

Bayer and Rebif by company Merck Sereno for multiple sclerosis have lost patent protection in

the United States in 2007 and most European Union countries in 2008. In a couple of years time

the pricing and sales of Betaferon (Betaseron) would be significantly affected due to the expiry

of the patents in Europe. Table 5.10 summarizes the currently available multiple sclerosis dugs

on market and patent expiry dates of these products. Fig 5.6 shows the predicted market share

of the multiple sclerosis drugs on market which would lose market share due to the entry of the

biosimilar version of the multiple sclerosis drug.

Fig 5.6: Predicted market share of multiple sclerosis drugs (2007-2017) Source: (Taylor, 2009).

Page 61: BIOSIMILARS: SCIENCE TO MARKET

52

Table 5.10: Multiple sclerosis drugs on the market and patent expiries. Source: Taylor P., 2009.

5.6.1.1.4 HUMAN INSULIN AND INSULINS ANALOGUES:

Insulin products such as Humulin and Novolin are used for the treatment of diabetes.

These insulin products are structurally identical to the naturally occurring insulin in human

pancreas. Later on insulin analogues were available in the market such as Lantus, Humalog,

NovoLog, Levemir and Apidra which are modified to make their properties better than natural

human insulin (Taylor, 2009). Table 5.11 shows that the original recombinant insulin products

such as Humulin and Novolin first launched in the market have lost their patent protection in

the year 2001 and 2002 respectively, but the insulin analogs which were introduced later will

expire during the 2013 to 2018 period.

Page 62: BIOSIMILARS: SCIENCE TO MARKET

53

Table 5.11: Recombinant insulin products on the market and patent expiries. Source: Taylor P., 2009.

5.6.1.1.5 MONOCLONAL ANTIBODIES (MAB):

Monoclonal Antibodies are an important category in the biopharmaceuticals consisting

of complex proteins which have a wide range of therapeutic applications such as cancer,

rheumatoid arthritis, asthma and psoriasis. Monoclonal Antibodies products have generated

sales revenue which is more than $21 billion in 2007. The Mab drug or product which first

succeeded commercially in the market and generated significant revenues was Rituxan as it

proved to be effective than most of the other therapies available on the market. Considering

the success of the Rituxan the drug developers launched various other Mab products such as

Avastin, Herceptin, Remicade, Humira and Erbitux. The innovator companies manufacturing

Mab are going to face competition from the biosimilar developers as major Mab products are

about to go off patent from 2012 (Taylor, 2009). Fig 5.7 illustrates the patent positions of the

leading biopharmaceutical products which are already expired or are about to lose patent

protection in the near future.

Page 63: BIOSIMILARS: SCIENCE TO MARKET

54

Fig 5.7: Estimated patent expiry dates of selected proteins Source: Taylor, 2009.

5.7 MARKET SHARE:

The current market share of the biosimilars contributes to only a small part of the sales

volume of the biologic drugs which are gone off patent. It has been reported that since 2005

only 25% of the biologic drugs have their patent status as expired and this indicates the

opportunity of more than $20 billion sales for biosimilars (Emmerich, 2010).

The Indian and Chinese biosimilar manufactures have launched more than 50 biosimilar

products in less regulated markets which is significantly high as compared to the US and

European manufactures (Emmerich, 2010). Fig 5.8 shows the market share for biosimilars in the

off patent biologics market. In the biologics market 23% of the biologic have gone off patent

and out of these 23%, the biosimilars market share is less than 5% which indicates the attractive

market opportunity for biosimilars to expand.

Page 64: BIOSIMILARS: SCIENCE TO MARKET

55

Fig 5.8. Market Share of biosimilars in the off patent biologics market. Source: Emmerich, R., 2010

Table 5.12 illustrates that the biosimilar market is quite small in the regulated markets

with less than 20 biosimilar products in the market.

Table 5.12: Bbiosimilar companies sales and market share. Source: Emmerich, 2010.

Page 65: BIOSIMILARS: SCIENCE TO MARKET

56

5.8 SALES:

Table 5.12 shows the global sales and the market share of the major biosimilar

companies which have launched biosimilar products. The market of the biosimilars is highly

fragmented and the global biosimilar companies represent only 15% of the market share.

Sandoz has the highest market share of 4.1% and global sales revenue of $23 million in the first

half of 2008(Emmerich, 2010). Sandoz has significant growth from 2007 due to the successful

authorization and introduction of Omnitrope which is a biosimilar human growth hormone and

Epoetin alpha hexal and Binocrit (epoetin products) in the European Union.

5.9 MARKET DRIVERS:

5.9.1 COST SAVINGS (GLOBAL HEALTH CARE):

The biopharmaceutical companies invest in huge amounts in the biologic product

development and manufacture due to the complex nature of the biologics as compared to the

investment involved in the development of traditional chemical generic. In 2010, the total

integrated sales of the top 12 biologic products were around $30 billion in the U.S. (Bourgoin,

2011). The Price of the biologic products indicates the development cost involved in biologic

products. The predicted standard cost of the biologic product per year is around $16,000 and

some of the other biologic products are more costly than the estimated price. For the treatment

of colorectal cancer, the therapies consisting of the biologic products can cost up to $10,000 per

month which is quite expensive (Bourgoin, 2011). In the U.S. and many other parts of the world

the government, federal and state initiatives like Medicare, Medicaid and NHS are responsible

for covering the expenses of these products. Therefore it is a great deal of interest to such

organizations, taxpayers and patients seeking for the prospect of cost savings through Biosimilar

products.

As compared to the chemical generics, the patient or the payer agency will not reap

significant savings in context with biosimilars but the savings could be appreciated when

Page 66: BIOSIMILARS: SCIENCE TO MARKET

57

compared to brand and it will be greatly substantial on individual level in terms of absolute

dollars.

The Generic Pharmaceutical Association (GPhA) made a statement in 2008 which

suggested that the health care system and the consumers could save billions of dollars if the

there is an availability of an effective route through which the patients have appropriate access

to harmless, effective and inexpensive biosimilar drugs (Taylor, 2009).

The pressure behind the uptake of the generic medicines and the requirement for the

biosimilar drugs is due to the increasing stress of cost containment in the major markets. The

potential savings possible or achievable by the usage of biosimilar drugs have been estimated

very differently in a variety of studies (Pandey, 2011).

It has been stated that if the biosimilars version of the most selling top 12 biologic drugs

whose patents are about to expire or have already expired are used for the period of ten years

then it could result in the savings of $67 billion to $108 billion. Further it has been added that

there would be savings of $236 billion to $378 billion if the same biosimilar version of that 12

biologic drugs are used for the period of twenty years (Taylor, 2009).

5.9.1.1 REGIONAL SAVINGS (U.S.)

The potential for the U.S. healthcare savings achievable through biosimilars for the time

period of 2009-2018 is estimated around $378 billion and it could be $ 2.0-2.8 billion over

period of ten years or $3.6 billion in 2008-2017 (Pandey, 2011). The passing of the biosimilars

legislation in the U.S. will enable cutbacks on total expenses on biologic medicines by $25 billion

over the time period of 2009-2018 which is recently estimated by the Congressional Budget

Office. The enactment of legislation will result in federal savings of more than $5.9 billion

including high returns of $0.8 billion and the competition will commence only after the first half

of the 2009-2018 periods and it will represent 0.5% of national expenses on total prescribed

medicines (Pandey, 2011).

Page 67: BIOSIMILARS: SCIENCE TO MARKET

58

A study carried out in the U.S. by the Pharmaceutical Care Manufacturers Association

(PCMA) estimated that on some biologics in the top 200 Medicare Part B reimbursed category

the federal savings could be in the range from $3.6 to $14.1 billion over the period of ten years

(Taylor, 2009).

It has also been reported that only the products from the four categories interferons,

erythropoietin, growth hormone, and insulin could help in the savings of $71 billion in the U.S.

alone over the period of ten years (Taylor, 2009).

It is stated in Reuter’s reports that in March 2010 legislation was passed to encourage

competition in the market of biologics. In the U.S. the Biologics Price Competition and

Innovation Act (BPCIA) authorized the sanction and marketing of the biosimilar products. It is

not anticipated that the biosimilars market will achieve same cost savings as the conventional

chemical generic drugs, but it is estimated that more than $300 billion amount of cost savings

could be possible by 2029 due to promotion of biosimilars. Reports also state that the

biosimilars products may cost up to 60 to 80 percent of the innovator biologic product

(Bourgoin, 2011).

Accessibility of the biosimilar version of the biopharmaceuticals will potentially save

billions of dollars of expenses of consumers annually and help preserve the solvency of the

Medicare program. As it is mentioned in Medicare report, by 2019 the funds available to the

Medicare trust will be exhausted if the Congress does not make any changes to legislation

(Taylor, 2009).

The prediction of cost savings obviously suggests that the cost containment strategy for the

global health care system would be the support and use of biosimilars. The healthcare

providers, payers and policy makers would be benefited greatly by the application and usage of

the biosimilars and cost containment strategy could be used by them in two ways:

Step therapy, which involves usage of the less expensive drugs before the usage or

recommendation of costly drugs.

Page 68: BIOSIMILARS: SCIENCE TO MARKET

59

Formulary alternative in which a plan is devised where the participants have access to some

medicines in a given category and participants make lowest co payment (Taylor, 2009).

5.9.1.2 REGIONAL SAVINGS (EUROPE):

The market dynamics were recently established in the European Union after the

regulatory framework was put into place.

The introduction of Gensulin which is recombinant insulin by Biotin in the Polish market

in 2001 resulted in savings of US$ 118 million in four years and savings in 2007 were up to US$

85 million (Sensabaugh, 2007). Omnitrope® is the biosimilar version of recombinant HGH which

was launched by Sandoz in Germany at a price lower than the novel reference product by 20

percent and in Australia the cost of the product will be about 25% less than the present

products of recombinant growth hormone. The discount on the biosimilar product is different

than the chemical generics which is generally 50-80 percent (Sensabaugh, 2007).

Page 69: BIOSIMILARS: SCIENCE TO MARKET

60

6. ISSUES & CHALLENGES:

6.1 COST OF PRODUCT DEVELOPMENT:

The cost of development of biosimilars is very unclear as different sources have

estimated a range of prediction levels for the development of particular biosimilar drug.

Generally the development cost is between $10 and $80 million but it has also been stated that

development of the simple biological drug could be between $14.5 million to $17 million as

compared to the chemical generic which is between $0.9 and $2.6 million (Clark, 2009).

Significantly higher estimates were presented by the CEO of TEVA in February 2009 which

indicated that developing a biosimilar would cost around $100 million to $150 million against $5

to $10 million for traditional chemical generic drugs (Clark, 2009). Regulatory pathways and

requirements play a crucial role in determining the cost of product development. In the

European Union, biosimilar applicants or biologic generic manufacturers & developers according

to regulations must prove the safety and efficacy of the biosimilar drug in the studies which can

cost up to $30 million to the developers. The companies who are looking forward to enter in

biosimilars competition in less regulated markets can expect the decreased development costs.

In less regulated biosimilar markets like India the development cost can be 90 percent less than

EU due to the lower regulatory standards faced by the biosimilar manufactures and developers

(Bourgoin, 2011).

In emerging, less regulated markets, the biosimilars manufactures are common as there

is low development cost which enables them to offer the biosimilar product at a much cheaper

price than the original innovator biologic product. Major generic companies may get attracted

to compete in less regulated emerging markets due to reasons like shorter time to market entry,

lower cost of development which enables these companies to recover investment for regulated

market competition. As compared to the standards of conventional generic industry the

development cost of biosimilars is significantly high, but this cost is relatively low when related

to the $1.2 billion investment which is required to develop a completely new Biologic drug

(Clark, 2009). A large amount of investment is consumed in clinical trials while developing a

Page 70: BIOSIMILARS: SCIENCE TO MARKET

61

Biosimilar drug. In developing biosimilars much of the savings in capital is achieved due to the

reduction of various stages like need for discovery, early stage “proof of concept” testing and

preclinical trials. Repetition of these steps would be redundant and unethical when the

efficiency of the innovators novel biologic is already established. From the company Hospira

management reports indicate that significant amount of savings can be achieved through the

capability of skipping phase II clinical trials when Hospira planned to apply for biosimilar

erythropoietin in EU (Clark, 2009). But companies cannot skip the large scale Phase III clinical

trials as it is essential to prove that biosimilar product has a similar safety and efficacy profile.

6.1.1 US REGION

The development cost of biosimilar drugs in the U.S. market will be the same in the EU

market if not more than that. There is a slight chance of additional higher development cost due

to additional supportive studies necessary for the approval especially for the developers looking

for interchangeability. In the U.S. market the biosimilar developers might also have to invest in

post marketing pharmacovigilance studies to survive the competition as these studies are vital

to gain approval from physicians and importantly the consuming patients. Pharmacovigilance

studies provide evident proof in distinguishing between biosimilars and the reference product

thus establishing physician confidence (Clark, 2009).

6.2 MANUFACTURING FACILITY:

It is estimated that it requires five to seven years time to bring the biological production

capacity online and it would cost around $300 million to $400 million to construct a plant which

is efficient in producing biosimilars (Clark, 2009). The outline of the Biosimilar companies is

exceptionally different as compared to that of the traditional generic companies due to various

reasons like involvement of the large investment capital funds and tedious regulatory hurdles.

Setting up of the manufacturing unit in the less regulated and emerging market like India or

china where tax and labour is cheap could decrease the time and expenditure but leads to other

augmented approval problems.

Page 71: BIOSIMILARS: SCIENCE TO MARKET

62

6.3 SUBSTITUTION:

Substitution or interchangeability includes the option between two or more drugs for

particular treatment area. Interchangeability between the original drug and biosimilar is outside

the remit of the evaluation conducted at the EMA; this aspect is not considered in the EU

guidelines. As the biosimilar is authorized it becomes optional treatment to others within the

same category of drugs. The substitution of the drugs depends on the regional policies of the EU

member states. In EU market the biosimilars products are distributed at hospitals which do not

come under retail pharmacy substitution policies. Several European countries such as Spain,

France and Germany have laws to stop automatic substitutions. For instance they have a list of

biologic products which cannot be automatically substituted. Therefore as per EMEA, treating

the patient with biosimilar or the innovator biologic product is responsibility of the qualified

healthcare professional or the internal policy followed at specific hospitals (Ruiz & Calvo, 2010).

According to the Hatch-Waxman Act the generic drugs possess the status of

interchangeability so the pharmacist can dispense generic even if the prescription was for

original drug. This makes possible for generic companies to gain market share without spending

capital on marketing. As per BPCIA if the biosimilar product gains the status as interchangeable,

then without the consent of the physician the biosimilar product can be substituted (Bourgoin,

2011).

6.4 MARKET EXCLUSIVITY:

The main objective of the data exclusivity is to guard the pharmaceutical registration

files. These registration files consist of the data which is generated and submitted to the

regulatory agencies like FDA and EMEA by the pharmaceutical companies during the process of

obtaining the marketing authorization of the new drug. Data exclusivity is different from patent

protection as it less restricted and any company can produce their own registration data.

However, due to the huge investment of funds and time required to produce this data inhibits

the generic companies.

Page 72: BIOSIMILARS: SCIENCE TO MARKET

63

In 2004 the European Union adopted the “8+2+1” formula for data exclusivity. As per the

formula the reference pharmaceutical product would have data exclusivity for eight years, two

years for market exclusivity and extra one year of protection for new indications of existing

products (Chu & Pugatch, 2009). In the two years of market exclusivity period the biosimilar

manufacturers will be able to submit the bioequivalence tests for their product or submit the

biosimilar applications. This data exclusivity formula almost equals to the 12 to 14 year period

which was suggested for biologics earlier. Fig 3.2 illustrates that the maximum market

exclusivity for the innovator reference product is 15 years.

In United States the topic of data exclusivity has received a lot of attention from various

investors. In the favor of companies, to recover from the huge investment and promote

innovation, the Biotechnology Industry Organization (BIO) insisted on the data exclusivity of 14

years as the breakeven point for the innovator drug is from 12.9 to 16.2 years of data

exclusivity. On the other hand Generic Pharmaceutical Association (GPHa) and White House

supported for the shorter time frame of about seven years. It has been reported by Bourgoin

(2011) that it is pointless to provide 12 to 14 years of exclusivity to stimulate innovation. The

patent protection for any product operate separately with that of market and data exclusivity

which implies that period of market exclusivity may end up doubtfully if the patent protection

surpasses the 12 year period which is adopted in BPCIA. Fig 6.1 shows the products which are

losing patent protection before 2020 and the time period of patent protection is greater than

that of market exclusivity period. The 12 year market exclusivity will protect the reference

product but this may lead the biosimilar applicants to follow the BLA process for approval as an

alternative to the abbreviated approval pathway.

Page 73: BIOSIMILARS: SCIENCE TO MARKET

64

Fig 6.1: Patent protection and market exclusivity for top biologics losing patent protection prior to 2018

Source: (Bourgoin, 2011)

There are also issues with overlapping of the process and the product patents along with

the exclusivity periods. These exclusivity periods inhibit the devolvement of biosimilar which is

fueled by the extension of patent protection which can be up to 5 years in some regions of the

world (Crandall, 2009).

6.5 INNOVATOR STRATEGIES:

There are number of ways by which the innovator companies of the biologic product

protect their intellectual property. Innovator companies are involved in influencing regulatory

bodies such as the FDA to be strict in developing a regulatory framework so that the biosimilar

product goes through the same costly and thorough testing as the original product. Another

Page 74: BIOSIMILARS: SCIENCE TO MARKET

65

fledging strategy which is employed by the innovator companies is called evergreening. This

strategy involves the extension of the patent protection by improving the product and launching

the improved version of the product resulting in increased outcomes. Table 6.1 illustrates the

modifications from the original product interferon to the pegylated interferons for the

treatment of hepatitis C resulted in extension of the patent protection. Another strategy which

is employed by giant companies is to license a generic product which is already authorized.

There is currently 12 years of exclusivity for the innovator and ambiguity in the legislation will

offer the innovator to apply for extra exclusivity of 12 years through the evergreening strategy.

This will result in innovator companies constantly making minor changes to the product and

obtain additional exclusivity which will restrain generic companies from development of the

biosimilar product. Citizen petition can also be filed by the innovator company on a generic

product prior to authorization of the generic drug; this will hamper the marketing of the product

as the citizen petition demands the issues to addressed associated with the product before it

enters the market. The innovators will strongly defend their product market share and revenues

by employing various tactics and one of the most common would be to convince medical

community the safety and reliability of the original innovator product as compared to the

biogeneric product (Crandall, 2009).

Table 6.1: Interferons on market and patent expiries. Source: Taylor, P. (2009)

Political lobbying by the pharmaceutical giants plays a crucial role and has a major

influence in the authorization of the biosimilars especially in the United States. It has been

estimated that the combined expenditure on political lobbying of various pharmaceutical,

Page 75: BIOSIMILARS: SCIENCE TO MARKET

66

healthcare devices and products in 2007 was up to $189million which was almost three times

the expenditure in 1998 which amounts to $67 million (Taylor, 2009). It has been reported by

Taylor (2010) that to defend the intellectual property rights, Amgen’s expenditure was about

$16.3 million, whereas the trade group Biotechnology Industry Organization and Pfizer’s

expenditure was about $7.2 million and $13.8 million respectively. The innovator companies

are also engaging into high profile law suits by taking legal action on any patent infringement to

block the generic manufacturers. For instance EPO is defended by Amgen from Genetic

institutes. The replacement of the original molecule by introduction of the second generation

products such as Aranesp was introduced by Amgen as the new improved version of Epogen.

Innovator companies opposing the biosimilar such as Amgen are urging various policy makers

on the labeling of the biosimilar product. Amgen suggests the label requirement stating the

clear difference between biosimilar and the brand product. To create further hurdles for

biosimilar developers, different common names should be allocated to the biosimilars and

clinical trials safety details should be provided on label is suggested by Amgen(Crandall, 2009).

6.6 PROFITABILITY OF BIOSIMILARS:

It has been reported by Biophoenix (2007) that the estimated cost to launch the

biosimilar product into market would be $21-55 million, keeping in mind the biomarker

endpoints can be used in clinical trials. For securing the production capacity involves a cost

which was more than $140 million for Sandoz. Biosimilars generally is not valid for automatic

substitution which leads to the additional investment in sales and marketing of the biosimilar

products. Biologics are usually dispensed from hospital specialists and rarely from the primary

physician, for instance EPO is generally sold to oncologist. In the case of the European Union for

the operation of the sales force of approximately 150 people along with infrastructure would

cost around $40 million. Most of the conventional generic companies do not have sales force.

Therefore to calculate the required sales to achieve considerable rate of interest is based on the

mentioned development and marketing cost. Thus a product with 10 years product life and 30

million development cost will require annual sales of €70million if sales force is to be set up

otherwise €40 million to achieve 70% gross margin.

Page 76: BIOSIMILARS: SCIENCE TO MARKET

67

The biosimilar market is also restricted as innovator companies have released

continuous second generation biologics which replaced original product as well as captured the

market share. The market has become congested as there are several innovators combined with

biosimilars players. As a result, the companies with availability of product development and

efficient sales & marketing infrastructure would have potential of growth in the emerging

biosimilar market. The growth in market could be achieved by various ways such as major

pharmaceutical companies with both innovative as well as biosimilar divisions like Novartis

consisting of Sandoz as the biosimilar development division. Another possibility is a strategic

partnership between the experienced biosimilar developers from Asia and generic experts from

Europe and USA.

6.7 MARKETING:

It has been reported that since the Hatch-Waxman (regulatory pathway for generic

drugs) it is more than 20 years and still 35% of the physicians do not consider that branded

drugs and their generic versions are equally effective. If this kind of view is observed for

conventional generic drugs then the biosimilars acceptance will face major difficulty. The

marketing model of the traditional generic drugs was based on the sale of the drugs to the

payers at relatively low cost as compared to the original branded drug and pharmacists entitled

to profits after the sale of the generic drug. But in the case of biologics, the administration of

the biologic is done through a physician. So, the Physician is the target of the biosimilar

manufacturers and the physician’s goal is to stress on clinical value more than economic gains.

The pharmaceutical giants will be in advantage such as Novartis who can lend marketing

expertise to Sandoz and Teva which has aggressive sales force supported by Copaxone. After the

introduction of Omnitrope by Sandoz in the European Union, it was reported that physicians

only recommends the biosimilar drug to the new patients and does not change the biologic drug

treatment of the patients which are already on the branded products. This situation arises due

to the marketing strategy used by the innovative company in which issues are raised against the

biosimilar product in terms of efficacy and safety (Clark, 2009).

Page 77: BIOSIMILARS: SCIENCE TO MARKET

68

7. CONCLUSION:

The Biosimilars are different from the traditional generic drugs so the regulations

applied to the generics are not applicable to the biosimilars. Chemical generics are the low

molecular weight compounds with a relatively straightforward process of production.

Manufacturing of the biosimilars is highly complex procedure due to their size and structure.

Biosimilar developers also have to face the challenges of maintaining the standards of safety,

efficacy and comparability with that of the reference product. The manufacturing of the

consistent product should be achieved by the biosimilar manufacturer as the manufacturing of

the biosimilar involves a series of steps which has to be conducted in controlled sequence and

the entire process has to be validated as the reproducibility of the biosimilar is difficult to

establish. The biosimilars are sensitive to the production process and manufacturing changes, as

a slight change in the process could result in change in the biological activity of the product

which will affect the clinical efficacy and safety.

The development of the biosimilars is a complicated task as it involves various issues

ranging from product development, to regulatory, to marketing and is a costly affair for the

pharmaceutical industry to bring it to reality. The present biopharmaceutical industry cost for

the development of a biosimilar and launching in the market is estimated to be around $100 to

$200 million (Pandey et al, 2011). On top of this the average period for the development of

biosimilar varies from 8 to 10 years which is roughly the time required to develop an original

novel biopharmaceutical product. Taking into account the current situation of the

pharmaceutical industry and the competition in the market, the developmental cost is

estimated to increase in the long term. Therefore many biopharmaceutical companies will take

chances of whether to choose between the research & development of the biosimilar or entirely

different new product.

The regulations should be designed in such a way that the market exclusivity periods

granted should encourage innovation. The regulations governing the biosimilars approval for

the market are complex. With the current regulatory framework it takes longer time for the

Page 78: BIOSIMILARS: SCIENCE TO MARKET

69

approval. For instance in Japan it takes about three years for approval. As the biosimilars are

more complex and different from the generics, the regulations for biosimilars are more

stringent than those of the generic drug approval. There are no synchronized regulations for

biosimilars which are followed all over the world but the European Union has the most

advanced and extensive regulatory framework for the market authorization of the biosimilars

and the rest of the world is about to follow the same standard. There are still no clear

definitions established in EMEA’s biosimilar development guidelines. For instance the biosimilar

approval decision can be influenced by the applicant ability to convince the Agency. Another

important criterion is that the biosimilar regulatory guideline should set up the requirements for

the clinical and non clinical data that are critical and sufficient to demonstrate biosimilarity. The

growth of the biosimilar market is restricted due to the uncertainties in regulatory framework

and complications arising in the clinical development and manufacturing.

According to the regulations the biosimilars are not granted automatic substitution

which means pharmacist cannot substitute the biosimilar with biologic. Due to lack of

experience and issues on the safety and efficacy of the biosimilars has lead to restrictions on

acceptance of biosimilars by physicians. Sales teams of the biosimilar companies have achieved

only limited success in promoting the usage of biosimilars and engaging in scientific

communications.

In the pharmaceutical market new dynamics can be introduced by the growth and

development of the biosimilars. Although there are various risks and challenges faced by the

biosimilar market, launch of biosimilars will open up attractive opportunities in the

pharmaceutical and generic sector. The uptake and demand for the biosimilars is increasing due

to the rising pressure of cost containment in the major markets. The potential healthcare cost

savings can be achieved due to the use of the biosimilars, as the prices of the biosimilars will

generally be 20% to 30% lower than innovator products. Apart from the lower prices on the

biosimilars, the main factor which will stimulate the growth of the biosimilar market is the

safety efficacy and convenience of the biosimilar products. Despite the strategies used by the

innovator companies, the competition in the future will not be between innovator companies

Page 79: BIOSIMILARS: SCIENCE TO MARKET

70

and generics but between second generation biologics based on advanced formulation or mode

of delivery or biologics with improved performance. Thus, emergence of the biologic will

indirectly help in the innovative research and with proper regulation will benefit patients,

healthcare system and industry.

Page 80: BIOSIMILARS: SCIENCE TO MARKET

71

REFERENCES:

Accenture (2009) Biosimilars- Emergence of the third market dynamic between original products and

generics. [Online] Available at:

http://www.biolawgics.com/Emergence%20of%20a%20Third%20Market%20Dynamic%20Between%20O

riginal%20Products%20and%20Generics.pdf 2 August 2011.

Borbosa, M. (2011). Immunogenicity of biotherapeutics in the context of developing biosimilars and

biobetters. Drug Discovery Today. Volume 16, Numbers7/8.April 2011.

Business insight Ltd, (2009) The Top 10 Biosimilar Players [Online] Available at:

http://www.biinteractive.com/index.aspx?StoryID=685486&ReportID=857&Lang=en&MainPage=render

Download Accessed on: November 19th 2010.

Bourgoin, A.F., (2011) WHAT YOU NEED TO KNOW ABOUT THE FOLLOW-ON BIOLOGIC MARKET IN THE

U.S.: IMPLICATIONS, STRATEGIES, AND IMPACT. London: Thomson Reuters. [Online] Available at:

http://thomsonreuters.com/content/science/pdf/ls/newport-biologics.pdf Accessed on: 2 August 2011.

Biophoenix (2007) Biosimilars, Biogenerics and Follow-on Biologics. London: Informa UK Ltd. [Online]

Available at:http://www.scripintelligence.com/multimedia/archive/00000/BS1342_117a.pdf Accessed

on: 2 August 2011.

Crandall, M (2009) The World Market For Biosimilars And The Future Of Follow-On Biologics. New York:

Kalorama Information.

Clark, T. D., (2009) Biosimilars: “PROBLEMS, PROSPECTS AND PROJECTIONS”. FLORIDA : VALUE OF

INSIGHT CONSULTING, INC., 2009. [Online] available at:

http://www.voiconsulting.com/freereports/Biosimilars%20Analysis%20Report.pdf Accessed on: 2 August

2011.

Crommelin, D., Teresa, B., Bissig, M., Damiaans, J., Krämer, J., Rambourg, P., Scroccaro, G., Strukelj, B.,

Tredree, R. (2005) Pharmaceutical evaluation of biosimilars: important differences from generic low-

molecular-weight pharmaceuticals. The European Journal of Hospital Pharmacy Science Volume 11 •

2005 • Issue 1 • P. 11 – 17.

Chu, R., Pugatch, M., (2009) Biogenerics or Biosimilars? Discussing the Present, Considering the Future.

[Online] available at: www.stockholm network.org/downloads/.../Biosimilars_FINAL.pdf Accessed on: 2

August 2011.

Chen, B. (2009) CMC Issues and Regulatory Requirements for Biosimilars. Trends in Bio/Pharmaceutical

Industry. Volume 5. Issue 4. 2009.

EuropaBio (2005) Biological and Biosimilar Medicines. [Online] available at:

www.europabio.org/documents/FS-Biosimilar.pdf Accessed on: 2 August 2011.

Page 81: BIOSIMILARS: SCIENCE TO MARKET

72

Evers, P. (2010) The Future of the Biologicals Market. Business Insights Ltd. [Online] available at:

http://360.datamonitor.com/Product?pid=BI00022-064&view=d0e11 Accessed on: 2 August 2011.

Emmerich, R. (2010) Origins and future of the biosimilars industry. Capgemini Consulting. [Online]

available at: www.dvfa.de/files/die_dvfa/.../life.../1_Ralf_Emmerich_Capgemini.pdf Accessed on: 2

August 2011.

EBE (2010) FDA REQUEST FOR COMMENTS ON “APPROVAL PATHWAY FOR BIOSIMILAR AND

INTERCHANGEABLE BIOLOGICAL PRODUCTS” *Online+ available at:

http://www.europabio.org/positions/Healthcare/EuropaBio_EBE_Joint_Submission_to_FDA_Final_21_1

2.pdf Accessed on: 2 August 2011.

Ehmann, F. (2010) Biosimilars –Regulation strategies and pathway in the EU (and US) Experience gained

and Perspectives. [Online] available at:

http://www.dvfa.de/files/die_dvfa/kommissionen/life_science/application/pdf/2_Falk_Ehmann_EMEA.p

df Accessed on: 2 August 2011.

EMA (2010) WORK PLAN FOR THE BIOSIMILAR MEDICINAL PRODUCTS WORKING PARTY (BMWP) 2011. .

[Online] available at:

http://www.ema.europa.eu/docs/en_GB/document_library/Work_programme/2009/11/WC500014447.

pdf Accessed on: 2 August 2011.

Fox, A. (2010) 'Biosimilar Medicines—New Challenges for a New Class of Medicine', Journal of

Biopharmaceutical Statistics, 20: 1, 3 — 9.

Greer, F.M. (2011) BIOSIMILAR DEVELOPMENT: FROM SCIENCE TO MARKET. Life science: Technical bulletin. Issue N°47 / JULY 2011. [Online] available at:

http://www.m-scan.com/media/1311266307692/Biosimilar-Development-From-Science-to-Market.pdf

Accessed on: 2 August 2011.

Horikawa, H., Tsubouchi, M., Kawakami, K. (2009) Industry views of biosimilar development in Japan.

Health Policy 91 (2009) 189–194.

Joshi, S.R. (2009) Biosimilar Insulins: Are they really ‘similar’ ? supplement of japi february 2009 vol.

57.38-39.

Kresse, G.-B. (2009) Biosimilars – Science, status, and strategic perspective. European Journal of

Pharmaceutics and Biopharmaceutics 72 (2009) 479–486.

Kox, S. (2009) The biosimilar framework in European Union. [Online] available at:

www.egagenerics.com/doc/jga_symposium_2009-02-18_skox.pdf Accessed on: 2 August 2011.

Marchant, J. (2007) The Future of Biosimilars. Business Insights Ltd [Online] available at:

http://360.datamonitor.com/Product?pid=BI00027-007 Accessed on: 2 August 2011.

Page 82: BIOSIMILARS: SCIENCE TO MARKET

73

Marre, M. & Kuhlmann, M., (2010) Lessons learned from biosimilar epoetins and insulins. British Journal

of Diabetes & Vascular Disease. 2010 10: 90.

Pandey, R. K., Srivastava, A.K., Rastogi, R., Mishra, S. K., (2011) Biosimilars: Overview of current

legislation and future prospects. Journal of Pharmacy Research Vol.4.Issue 4. April 2011.

Ruiz, S., Calvo, G. (2010) Similar biological medicinal products: lessons learned and challenges ahead. .

Journal of Generic Medicines: The Business Journal for the Generic Medicines Sector. Volume 8,1,4-13.

Sensabaugh, S.M. (2007) Biological generics: A business case. Journal of Generic Medicines: The Business

Journal for the Generic Medicines Sector 2007 4: 186.

Sharma, B. (2007) Manufacturing challenges for biosimilars- The process defines the product. EJHP

Practice - Vol.13 2007/Issue 5.

Sauer, T. and Kramer, I. (2010) The new world of biosimilars: what diabetologists need to know about

biosimilar insulins. British Journal of Vascular Disease 2010 10:163.

Taylor, P. (2009) Negotiating the Emerging Biosimilars Landscape. Business Insights Ltd. [Online]

available at: http://360.datamonitor.com/Product?pid=BI00027-018&view=d0e11 Accessed on: 2 August

2011.

Wang, S. (2011) Patent search on biologics as potential biosimilar candidates. World Patent Information

33 (2011) 67–71.

Zuniga, L., Calvo, B., (2009) Regulatory aspects of biosimilars in Europe. Trends in Biotechnology Vol.27

No.7.

Zuniga, L., Calvo, B. (2010) Biosimilars approval process. Regulatory Toxicology and Pharmacology 56

(2010) 374-377.