developing oncology biosimilars: an essential approach for the future

20
Developing Oncology Biosimilars: An Essential Approach for the Future Jame Abraham The treatment of many diseases, particularly cancer, has been profoundly impacted by the introduction of biologic therapies (biologics), which are incorporated into the treatment algorithms of most oncology clinical practice guidelines. Biologics are large molecular weight, structurally complex proteins that are produced via complex manufacturing processes. With the patents of key biologics, including many widely used in oncology, set to expire in the near future, a number of pharmaceutical companies have focused on developing biosimilars. While the goal of development is to demonstrate that the biosimilar product is highly similar to the reference biologic product, biosimilars should not be viewed as genericbiologics. Generic drugs are small chemical moieties that are identical to the patent-expired referencesmall- molecule drugs. It is not possible to produce an identical copy of a biologic, so the term biosimilarwas chosen to define an appropriately similar biologic product. Improving patient access to cancer therapies such as biologics and reducing healthcare costs are key initiatives of the US Government; the integration of approved biosimilars into clinical practice will be instrumental in accomplishing these goals. Semin Oncol 40 (Suppl 1):S5-S24 & 2013 Elsevier Inc. All rights reserved. B iologic drugs (biologics) are produced in living biological systems, whereas small- molecule drugs (eg, aspirin, statins, and ben- zodiazepines) are manufactured using standard chemical synthesis procedures. 1 The methods used to develop most small-molecule drugs are relatively easy to replicate, which facilitates the eventual availability of generics upon patent expiration. 2 In contrast, the multifaceted manufacturing processes required to produce biologics are rendered even more complex by the high level of structure asso- ciated with functional proteins. 3 Due to these com- plexities, not to mention the proprietary nature of manufacturing processes, competing pharmaceutical companies are unable to exactly duplicate approved biologics. These issues have resulted in the develop- ment of biosimilars.2 Biosimilars are considered as highly similarbut not identical to their reference biologic products (ie, they should not be viewed as biogenerics). 4 As defined by federal law (section 351(i) of the Public Health Service [PHS] Act), the term biosimilarrefers to a biologic product that is highly similar to the reference biologic product, notwithstanding minor differences in clinically inactive compo- nents.5 There should be no clinically meaningful differences between biosimilars and reference bio- logics in terms of safety, purity, and potency (Table 1). 6,7 Unlike small-molecule generic drugs, biosimilars are structurally complex, large molecular weight proteins that are subject to a variety of post- translational modifications. 2,810 To increase patient access to cancer therapies, more cost-effective biosimilars may become available in the United States upon patent expiration of their reference biologic products. 2,6,11,12 However, the anticipated entry of oncology biosimilars into the US market in the next few years creates a unique set of challenges for stakeholders (eg, the pharmaceut- ical companies involved in the development of biosimilars), as well as legal and regulatory author- ities, healthcare providers (HCPs), and patients. The US regulatory and approval processes for small- molecule generic drugs are not applicable to 0093-7754/ - see front matter & 2013 Elsevier Inc. All rights reserved. http://dx.doi.org/10.1053/j.seminoncol.2013.09.015 This supplement was funded by Pfizer Inc. Statement of conflict of interest: Jame Abraham received no financial support from Pfizer Inc for the research and/or authorship of this supplement. Medical writing and editorial support to prepare this sup- plement was provided by Joseph Abrajano, PhD, and Stephen Strudwick, PhD, of QD Healthcare Group and funded by Pfizer Inc. Director of Breast Oncology Program, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH. Address correspondence to Jame Abraham, MD, FACP, Taussig Can- cer Institute, Cleveland Clinic, 9500 Euclid Ave R-35, Cleveland, OH 44195. E-mail: [email protected] Seminars in Oncology, Vol 40, No 6, Suppl 1, December 2013, pp S5-S24 S5

Upload: jame

Post on 27-Jan-2017

216 views

Category:

Documents


1 download

TRANSCRIPT

Page 1: Developing Oncology Biosimilars: An Essential Approach for the Future

Developing Oncology Biosimilars: An Essential Approachfor the Future

Jame Abraham

The treatment of many diseases, particularly cancer, has been profoundly impacted by the

introduction of biolo

algorithms of most on

structurally complex pthe patents of key biol

future, a number of ph

the goal of developmereference biologic pro

drugs are small chem

molecule drugs. It is“biosimilar” was chose

access to cancer thera

the US Government;instrumental in accom

Semin Oncol 40 (Supp

0093-7754/& 2013 Elshttp://dx.doi

This suppleStatement osupport frosupplementplement wStrudwick,

Director oClevelan

Address cocer InstitOH 4419

Seminars

gic therapies (biologics), which are incorporated into the treatment

cology clinical practice guidelines. Biologics are large molecular weight,

roteins that are produced via complex manufacturing processes. Withogics, including many widely used in oncology, set to expire in the near

armaceutical companies have focused on developing biosimilars. While

nt is to demonstrate that the biosimilar product is highly similar to theduct, biosimilars should not be viewed as “generic” biologics. Genericical moieties that are identical to the patent-expired “reference” small-

not possible to produce an identical copy of a biologic, so the termn to define an appropriately similar biologic product. Improving patient

pies such as biologics and reducing healthcare costs are key initiatives of

the integration of approved biosimilars into clinical practice will beplishing these goals.

l 1):S5-S24 & 2013 Elsevier Inc. All rights reserved.

Biologic drugs (biologics) are produced in

living biological systems, whereas small-

molecule drugs (eg, aspirin, statins, and ben-zodiazepines) are manufactured using standard

chemical synthesis procedures.1 The methods used

to develop most small-molecule drugs are relativelyeasy to replicate, which facilitates the eventual

availability of generics upon patent expiration.2 In

contrast, the multifaceted manufacturing processesrequired to produce biologics are rendered even

more complex by the high level of structure asso-

ciated with functional proteins.3 Due to these com-plexities, not to mention the proprietary nature of

manufacturing processes, competing pharmaceutical

companies are unable to exactly duplicate approved

- see front matterevier Inc. All rights reserved..org/10.1053/j.seminoncol.2013.09.015

ment was funded by Pfizer Inc.f conflict of interest: Jame Abraham received no financialm Pfizer Inc for the research and/or authorship of this. Medical writing and editorial support to prepare this sup-as provided by Joseph Abrajano, PhD, and StephenPhD, of QD Healthcare Group and funded by Pfizer Inc.

f Breast Oncology Program, Taussig Cancer Institute,d Clinic, Cleveland, OH.

rrespondence to Jame Abraham, MD, FACP, Taussig Can-ute, Cleveland Clinic, 9500 Euclid Ave R-35, Cleveland,5. E-mail: [email protected]

in Oncology, Vol 40, No 6, Suppl 1, December 2013,

biologics. These issues have resulted in the develop-

ment of “biosimilars.”2

Biosimilars are considered as “highly similar” butnot identical to their reference biologic products

(ie, they should not be viewed as “biogenerics”).4 Asdefined by federal law (section 351(i) of the PublicHealth Service [PHS] Act), the term “biosimilar”refers to a “biologic product that is highly similar

to the reference biologic product, notwithstandingminor differences in clinically inactive compo-

nents.”5 There should be no clinically meaningful

differences between biosimilars and reference bio-logics in terms of safety, purity, and potency

(Table 1).6,7 Unlike small-molecule generic drugs,

biosimilars are structurally complex, large molecularweight proteins that are subject to a variety of post-

translational modifications.2,8–10

To increase patient access to cancer therapies,

more cost-effective biosimilars may become available

in the United States upon patent expiration of theirreference biologic products.2,6,11,12 However, the

anticipated entry of oncology biosimilars into the

US market in the next few years creates a unique set

of challenges for stakeholders (eg, the pharmaceut-

ical companies involved in the development of

biosimilars), as well as legal and regulatory author-

ities, healthcare providers (HCPs), and patients. The

US regulatory and approval processes for small-

molecule generic drugs are not applicable to

pp S5-S24 S5

Page 2: Developing Oncology Biosimilars: An Essential Approach for the Future

Table 1. FDA Definitions of Biologics, Biosimilars, and Generic Drugs

Biologicalproducts

� The term ‘‘biological product’’ means a virus, therapeutic serum, toxin, antitoxin,vaccine, blood, blood component or derivative, allergenic product, protein (exceptany chemically synthesized polypeptide), or analogous product, or arsphenamine orderivative of arsphenamine (or any other trivalent organic arsenic compound)applicable to the prevention, treatment, or cure of a disease or condition of humanbeings

Biosimilars � The term ‘‘biosimilar’’ or ‘‘biosimilarity,’’ in reference to a biological product that isthe subject of an application under subsection (k), means:(A) The biological product is highly similar to the reference biologic product

notwithstanding minor differences in clinically inactive components(B) There are no clinically meaningful differences between the biological product

and the reference biologic product in terms of the safety, purity, and potency ofthe product

Generic drugproducts

� A generic drug is the same as a brand name drug in dosage, safety, strength, how itis taken, quality, performance, and intended use. Before approving a generic drugproduct, the FDA requires many rigorous tests and procedures to assure that thegeneric drug can be substituted for the brand name drug. The FDA bases evaluationsof substitutability, or therapeutic equivalence, of generic drugs on scientificevaluations. By law, a generic drug product must contain the identical amounts ofthe same active ingredient(s) as the brand name product. Drug products evaluatedas therapeutically equivalent can be expected to have equal effect and no differencewhen substituted for the brand name product

US House of Representatives,5 Generics and Biosimilar Initiative.6

J. AbrahamS6

biosimilars. Some key questions to be discussed in

this article include the necessity of safety surveil-

lance (pharmacovigilance) procedures, the appropri-ateness of interchangeability and substitution

practices, and the need for appropriate nomencla-

ture for biosimilars.Because all biologic drugs (ie, biosimilars and

their reference biologic products) are proteins, they

have the potential to induce antibody responses inpatients, which may result in hypersensitivity reac-

tions and/or other adverse events (AEs). The immu-

nogenic potentials and AE profiles of biologics arealways assessed in clinical trials and monitored post-

approval. Biosimilars are not identical to the licensed

reference biologic products, thus the safety ofbiosimilars up to and postapproval needs to be

monitored closely (eg, through postmarketing phar-

macovigilance procedures).Despite these considerations, the hope is that the

US Food and Drug Administration (FDA) approval

process for biosimilars will be abbreviated (com-pared with the approval process for a new biologic

drug application), with biosimilars effectively pro-

duced at lower costs than their reference biologicproducts.2,13 Although specific requirements for the

demonstration of appropriate similarity between

biosimilars and their reference biologic productshave not been fully defined by the FDA, these

requirements likely will be tailored for each of the

different types of biologics. For example, the

requirements for similarity for very structurally com-plex protein molecules such as monoclonal anti-

bodies (mAbs) will likely be more extensive than

those for biologics with simpler structures (eg,somatotropin). Not surprisingly, the FDA most likely

will consider applications on a case-by-case basis

using a review process that considers the “totality ofthe evidence.”2,14

Biosimilars are expected to increase global patient

access to therapies for a range of diseases includingcancer. The integration of biosimilars into clinical

practice has already begun in countries including

those in the European Union (EU). In 2005, theEuropean Medicines Agency (EMA) issued general

guidelines on biosimilars, and the European Commis-

sion subsequently implemented revised pharmaceut-ical legislation, which established the first formal

regulatory framework for the evaluation, authoriza-

tion, and monitoring of biosimilars.13–17 The refer-ence biologic product for the applicant biosimilar

must be registered in the EU, and the applicant

biosimilar must be similar in pharmaceutical form,mode of administration, and strength to the reference

biologic product.13,17 A biosimilar for somatotropin

was approved in the EU in 2006, followed bybiosimilars for erythropoietin and filgrastim.

Page 3: Developing Oncology Biosimilars: An Essential Approach for the Future

Developing oncology biosimilars S7

Key understandings from the integration of biosimi-

lars into healthcare practices in the EU and other largeand emerging markets (eg, Brazil, China, and India) are

discussed in this review.18 The federal government, and

biotechnology and pharmaceutical industries shouldlearn from global experiences with biosimilars and

develop effective manufacturing and approval proc-

esses to facilitate the safe integration of biosimilars intoUS clinical practice. In 2012, the FDA took a significant

step toward this goal by releasing draft guidance on the

development and approval of biosimilars.19

INTRODUCING BIOSIMILARS TO EXPANDPATIENT ACCESS TO CARE

Biologic drugs have had a significant impact on all

areas of medicine, most notably in the fields ofoncology and rheumatology. Biologics are included

in treatment guidelines such as those of the National

Comprehensive Cancer Network (NCCN).1 Examplesof biologics that have had a significant impact on the

practice of oncology include rituximab (FDA

approved for the treatment of diffuse large B-cellnon-Hodgkin’s lymphoma) and trastuzumab (FDA

approved for the treatment of HER2-overexpressing

breast cancer, and metastatic gastric or gastroesopha-geal junction adenocarcinoma). Biologics also play an

active role in other types of cancers including colon,

head and neck, kidney, non–small-cell lung cancers,and chronic lymphocytic leukemia.1 The driving

forces behind the development of oncology biosimi-

lars include expanding patient access to these effec-tive biologic therapies, which has been shown to be

an important predictor of survival by the American

Cancer Society, and reducing healthcare costs.12,20

The US Hatch-Waxman Act of 1984 enhanced

patient accessibility to small-molecule drugs and

increased affordability by allowing abbreviatedapproval of thousands of small-molecule generic

(ie, nonbiologic) drugs following patent expiration

of the small-molecule reference products.21,22 Inmuch the same way, the Biologics Price Competition

and Innovation (BPCI) Act of 2009, a component of

Epoetin alfaFilgrastim PegUnited

States

Europe RituximabCetuximabInfliximab

Trastuzumab

2013 2014

Figure 1. Patents for many biologics are set to expire in

the Patient Protection and Affordable Care Act

(PPACA) signed into US law in 2010, was enactedwith the goal of expanding patient accessibility by

allowing price competition among biologics and

reducing costs. Such legislation has encouragedpharmaceutical and biotechnology companies to

develop biosimilars for use in the United States,

especially as the patents of several key biologics,including several widely used in oncology, are set to

expire in the near future (Figure 1).17,23–25

The use of biosimilars in the EU is beginning toshow evidence of healthcare improvements predicted

to arise from expanding patient access to effective

biologic therapy.3,11 The EU introduction of a biosimilarfor the biologic reference product filgrastim has been

accompanied by a trend towards increased overall use

of filgrastim as prophylaxis against primary neutrope-nia, which has been effective in preventing chemo-

therapy dose reductions and discontinuations.3,11 In the

United Kingdom, the launch of a biosimilar of filgrastimhas reduced per patient spending and increased access

for patients. Specifically, while spending rose only 38%,

filgrastim volume more than doubled from Q1 2008 toQ2 2012. The improved healthcare afforded by the

introduction of this biosimilar may relate in part to the

decision by physicians to preferentially use it earlier inthe course of disease, given its lower cost versus the

reference biologic product. The cost-savings afforded

by using biosimilars in supportive care settings (eg,biosimilars for the biologic reference products of

filgrastim and erythropoietin) may allow more patients

to use life-saving biologic therapies such as rituxi-mab.3,11 The incorporation of biosimilars into US

oncology clinical practice is likely to significantly

impact the cost of cancer therapy.

EXPANDING PATIENT ACCESS TO CARE BYREDUCING HEALTHCARE COSTS

Of the nearly $287 billion spent by Americans

on prescription drugs in 2007, approximately $40billion was attributable to biologics.20,21 Recent data

indicate that 60% of expenditures for the top 10

filgrastimAdalimumabCetuximab

BevacizumabDarbepoetin

InfliximabTrastuzumabRituximab

DarbepoetinAdalimumabBevacizumabPegfilgrastim

2015 2016 2017+

the near future. Generics and Biosimilar Initiative.25

Page 4: Developing Oncology Biosimilars: An Essential Approach for the Future

37.7%

62.7%

Nonbiologics$2785

Biologics$4610

Figure 2. Distribution of drug expenditures for the top10 antineoplastic drugs in outpatient clinics (in millionsof dollars). Zelenetz et al,1 Doloresco et al.26

J. AbrahamS8

antineoplastic drugs in 2010 were attributed to theuse of biologics (Figure 2).1,26

The biologic drugs bevacizumab, rituximab, and

trastuzumab were the top three antineoplastic drugsused in 2010, and they collectively accounted for

approximately 58% of the top 10 total antineoplastic

drug expenditures (Table 2).1,26 By comparison, thetop three nonbiologic antineoplastic drugs (doce-

taxel, pemetrexed, and oxaliplatin) accounted for

approximately 24% of the total expenditures.1,26

The eventual integration of biosimilars into oncol-

ogy treatment regimens may significantly impact

patient care in the United States, especially as thecost of biologics is a limiting factor for many

patients. The introduction of biosimilars in the EU

may be resulting in expanded access to life-savingtherapies and use of effective therapies much earlier

in the course of disease.3,11 Given the complex,

multifaceted manufacturing processes required toproduce biologics, it is estimated that biosimilars

will cost between at least 15% and 30% less than the

Table 2. Top 10 Antineoplastic Drug Expenditures i

Rank Drug Type of Drug2010 Exp

($ Mi

1 Bevacizumab Biologic 182 Rituximab Biologic 143 Trastuzumab Biologic 94 Docetaxel Nonbiologic 65 Pemetrexed Nonbiologic 56 Oxaliplatin Nonbiologic 57 Gemcitabine Nonbiologic 48 Cetuximab Biologic 39 Bortezomib Nonbiologic 3

10 Leuprolide Nonbiologic 2Total (Top 10) — 73

Zelenetz et al1 and Doloresco et al.26

reference biologic products. In the EU, biosimilar

versions of recombinant human erythropoietin(rhEPO) have been reported to be 25% to 30% lower

in cost than the reference biologic products. In

comparison, small-molecule generic drugs lowercost up to 90% compared with the small-molecule

reference products.27

In terms of the EU economy, it is too soon toevaluate the impact of biosimilars on the biologics

market. It is important to note, however, that some

EU manufacturers of reference biologic productshave lowered their prices in response to the intro-

duction of biosimilars, which will complicate the

assessment of the economic impact of biosimilarsin the EU and may cause a decrease in interest in

switching to biosimilars.13,27

BACK TO BASICS: COMPARING BIOLOGICSWITH SMALL-MOLECULE DRUGS

For the purposes of this discussion, a “small-molecule drug” refers to any chemically synthesized

drug for which a generic version can be produced

easily (eg, statins and benzodiazepines). Biologics arelarger molecules with many more levels of complex-

ity than small-molecule drugs (Table 3).2,7–10,23,28–31

For example, aspirin is a small-molecule drug with

a well-defined and homogeneous chemical structure;

by comparison, biologics such as trastuzumab maybe between 100 to 1,000 times larger in molecular

weight and are more structurally complex

(Figure 3).8,9,32,33

All biologic drugs are proteins and as such consist

of a defined sequence of amino acids, which con-

stitute the primary structure.1,9 While the primarystructure is essential for biologic activity, additional

n Outpatient Clinics

endituresllions)

Percent of TotalExpenditures

84 26% Top 366 20% Biologics31 13% 59%88 9% Top 379 8% Nonbiologics08 7% 24%63 6%29 4%27 4%20 3%95 100%

Page 5: Developing Oncology Biosimilars: An Essential Approach for the Future

Table 3. Key Differences Between Biologics and Conventional Small-Molecule Drugs

Difference Small-Molecule Drugs Biologic Drugs

Size Generally small molecules � Large, complex, protein/glycoproteinmolecules

Structure Simple, noncomplex structure; impurities/contaminants can be easily assessed inrelation to the reference biologic product

� Multiple levels of structure (primary,secondary, tertiary, quaternary) as well aspost-translational modifications; exactstructure cannot be easily assessed inrelation to the reference biologic product

Forms Typically only 1 form and homogeneous � Heterogeneous; multiple distinct isoformsmay be present in a given preparation

Immunogenicity Generally immunologically inert � Potentially immunogenic;immunogenicity versus the referencebiologic product cannot be fullyevaluated

Manufacturingprocess

Produced via chemical synthesis;manufacturing process of the referenceproduct can easily be replicated

� Produced in living systems; manufacturingprocess of the reference biologic productis typically proprietary and cannot beexactly replicated

Stability Typically stable and predictable � May be exquisitely sensitive to physicalconditions (eg, storage conditions, pH,temperature, agitation)

Declerck,2 Cai et al,7 Lee et al,8 Kuhlmann et al,9 Kanter et al,10 Mellstedt et al,23 Nowicki,28 Rathore et al,29 Vulto et al,30 EMA.31

Developing oncology biosimilars S9

levels of structure, including secondary (ie, protein

folding due to hydrogen bonding), tertiary (ie, associ-ations between secondary structures) and quaternary

(ie, associations between two or more protein chains)

structures, also may be essential to confer the ther-apeutic and even the toxic effects of a biologic

(Figure 4).1,10 In addition, the functionality of a

biologic drug may be altered by a wide range ofpost-translational modifications, such as glycosylation,

methylation, acetylation, phosphorylation, and/or

hydroxylation.2

Figure 3. Compared with small-molecule drugs, biologic drugs aversion of this article at seminoncol.org for color figure. (Note: figur

The term “generic” should not be associated with

biosimilars, and should be used to refer to identicalcopies of small-molecule drugs that possess

precisely-defined atomic structures.9,23 In general,

small-molecule generic drugs are identical to thereference small-molecule drugs and can be synthe-

sized reliably and consistently (Table 3). As a result,

the manufacturing, marketing, and integration ofsmall-molecule generic drugs into clinical practice

has been relatively straightforward. A manufacturer

of a small-molecule generic drug needs to demon-

re larger and more complex in structure. Refer to the webe is for illustrative purposes and not intended to be to scale.)

Page 6: Developing Oncology Biosimilars: An Essential Approach for the Future

N

Tertiary

C

N N

Quaternary

NN

CC

C C

SecondaryPrimary

F

L

KD

F L

LV T

T S

S

VT P A

F A H

A

S V

K Y

Figure 4. Levels of protein structure (ie, for biosimilars and the reference biologic products). Refer to the web version ofthis article at seminoncol.org for color figure.

J. AbrahamS10

strate physiochemical identity with the reference

biologic product and pharmacokinetic equivalence;the approval process typically requires proof of

quality and pharmacokinetic bioequivalence to the

reference biologic product, and no substantive clin-ical data.9

While biosimilars can be considered to be “sim-

ilar” to the reference biologic products, they cannotbe deemed “identical” due to the inherent complex-

ity of proteins (Table 3).1,17 Differences in any

structural characteristics (eg, more or less glycosyla-tion of a biosimilar versus its reference biologic

product) may potentially impact the efficacy of the

biosimilar, and may increase the potential for AEs.7

Even in the event that all levels of structure for a

given reference biologic product can be reproduced

sufficiently and the biosimilar shows similar activity,the potential still exists for some minute differences

to go undetected, which may result in unforeseen

AEs.10 In addition, even slight differences in abiosimilar (compared with the reference biologic

product) could be sufficient to stimulate an immune

response (eg, a hypersensitivity reaction) not typi-cally observed with the reference biologic prod-

uct.2,10 Finally, even if a biosimilar uses the same

cell lines, reagents, and methodologies as those usedby the reference biologic product, it may not be

possible to completely replicate the exact cell

culture fermentation conditions needed to producethe reference biologic product.7 Because the activity

of biologics is highly dependent on the manufactur-

ing conditions and process, this can lead to productheterogeneity and differences in activity between a

biosimilar and a reference biologic product that can

potentially extend to different manufactured batchesor lots of the biosimilar.1,9

Because biosimilars are different from small-

molecule generic drugs, the manufacturing proc-esses are different, and the regulatory and safety

surveillance (ie, pharmacovigilance) processes that

ultimately are implemented in the United States willbe specific to biosimilars. The United States will

likely follow the lead of the EMA, which authorizes

the use of biologics, whereas use of chemically-derived medicines can be authorized by the member

country’s regulatory agency.13 The regulatory appro-

val pathway for biosimilars has not been fullydefined by the FDA,1 but based on initial draft

guidance, this approval pathway will require a

stepwise “totality of evidence” approach, wherebythe extent of the data provided may be variable

based on type of biosimilar under consideration (to

be further discussed later in this review).19,34

THE BIOSIMILAR MANUFACTURING PROCESS

The World Health Organization (WHO) has sug-

gested that the manufacturing process for bio-

similars be designed to minimize the differencesbetween the biosimilar and the reference biologic

product. The goal of minimizing differences is to

reduce the clinical testing requirements for thebiosimilar by referring to as much of the clinical

history of the reference biologic product as possi-

ble, while decreasing any predictable impact onclinical safety and efficacy.35 Initial guidance from

the FDA has recommended that manufacturers

follow the principles outlined within the Interna-tional Conference on Harmonization (ICH) Q5E

guidelines relating to any change in the manufactur-

ing process for a marketed protein product to “assessthe effects of the [manufacturing] change and demonstratethrough appropriate analytical testing, functional assays,and/or in some cases animal and/or clinical studies, that thechange does not have an adverse effect on the identity,strength, quality, purity, or potency of the product as theyrelate to the safety or effectiveness of the product.”19 None-theless, because the manufacturer of the biosimilar

will be different from the manufacturer of the

reference biologic product, additional data beyondthat required in ICH Q5E guidelines will be needed

to effectively demonstrate biosimilarity.19 Accord-

ingly, a major technical challenge in manufacturingbiosimilars is the knowledge gap caused by the fact

Page 7: Developing Oncology Biosimilars: An Essential Approach for the Future

Figure 5. Manufacturing process for biosimilars. Refer to the web version of this article at seminoncol.org for colorfigures. Black and white images: Kuhlmann M, Covic A. The protein science of biosimilars. Nephrol Dial Transplant.2006;21(Suppl 5):v4–8,9 by permission of Oxford University Press. Color images: Iculig/Shutterstock.com; Webspark/Shutterstock.com; Dr Gopal Murti/Science Photo Library; Jennifer Waters/Photo Researchers.

Developing oncology biosimilars S11

that the process for developing and manufacturingthe reference biologic is proprietary.8,23

The key steps involved in producing a biologic are

outlined in Figure 5.9,23,28 These steps includeexpressing the protein in the most appropriate cell

line, producing protein at a commercial level, down-

stream processing, and purification.9,23,28 The cellculture process required for producing a biologic

is considered especially critical in determining identity,

purity, and potency of the finished product.8 Specif-ically, the manufacturing process generally begins

with the gene sequence of the protein of interest

being cloned into an expression vector that is thentransfected and expressed in a unique cell line

derived from a single clonal cell.28 The cell line

expressing the protein must then be rigorouslyevaluated for a number of performance parameters,

including integrity and quality, as well as growth and

viability under the required manufacturing condi-tions.28 Several additional factors can impact the

quality of a manufactured biologic drug, including

source/batch of any of the required raw materials,conditions (eg, temperature, pH, light exposure, and

agitation), chemical substances that can leach from

storage containers, and the equipment used to massproduce the product.8,29

Some of the stages in the biosimilar manufactur-

ing process where potentially significant differen-ces from the manufacturing process of the refer-

ence biologic product may emerge are outlined in

Figure 6.9,28,36 The increased sensitivity of biologicsto changes in physical conditions necessitates strict

control not only over the manufacturing process,but also over the storage and administration con-

ditions for the finished product.8,28 Unlike chemi-

cally synthesized small-molecule drugs that aremore stable and have predictable characteristics,

biologics can be more sensitive to changes in

physical storage conditions of the final product(eg, temperature and pH). Finally, the conditions

in which biosimilars are administered can affect

the final product. For example, whereas most small-molecule drugs can be taken orally, many biologics

must be either injected or inhaled as they are

more sensitive to enzymatic degradation in the dig-estive tract.28

Special Considerations: Biosimilar MonoclonalAntibodies

The top three oncology biologics (bevacizumab,rituximab, trastuzumab) in terms of the expenditures

of outpatient clinics are mAbs (Table 2).26 The

manufacturing process for mAbs can be compli-cated, as mAbs may exist in different structural

isoforms or structural variants, and their activity may

be sensitive to factors such as glycosylation or otherpost-translational modifications.8 The predominance

of a given isoform or variant and/or the presence of

other isoforms or variants in a specific biosimilarpreparation versus its reference biologic product

should be characterized, as this could significantly

impact the efficacy and safety, as well as immuno-genicity of the mAb.8,37

Page 8: Developing Oncology Biosimilars: An Essential Approach for the Future

Cell expansion

Cell production in bioreactors

Recovery throughfiltration or

centrifugation

Purification throughchromatography

Characterizationand stability

Different cell line,growth media,

method of expansion

Different cell line,growth media,

bioreactor conditions

Different operatingconditions

Different binding and elution conditions

Different methods,reagents, reference

standards

PurifiedBulk Drug

Figure 6. Manufacturing biosimilars: differences from the reference biologic product may occur at multiple steps in theprocess. Mellstedt et al.23

J. AbrahamS12

In addition to “magic bullet” type specificity of

antigen binding, mAbs have fragment-crystallizable(Fc) regions that can mediate a variety of important

effector functions, such as antibody-dependent cell

cytotoxicity (ADCC) and complement dependentcytotoxicity (CDC), which may be related to their

therapeutic effects.2 Therefore, even if antigen bind-

ing is believed to be the primary mechanism ofaction, it cannot be assumed that Fc effector func-

tions are clinically irrelevant.38 The type and level

glycosylation of the Fc portion of a mAb has thepotential to impact some of the effector functions of

mAbs, such as ADCC and CDC, as well as their

clearance from the body.2 Accordingly, when devel-oping and testing biosimilar mAbs, it will be impor-

tant to evaluate not only the major antibody binding

characteristics, but also the effector functions.Extensive in vitro studies should be conducted to

assess activities such as target antigen binding,

Table 4. Potential Challenges When Bringing Biosim

Challenge

Complex production and analysis procedures,relative to other non-mAb biosimilar products

� Hsa

Competing development of next-generationmAbs with improved stability and/or efficacy

� Sgg

Competing emergence of other antibody-therapeutic technologies (eg, bi-specific mAbs,Fc-linked fusion molecules)

� Sg

Competing emergence of cheaper antibody-producing methodologies (eg, nanoantibodiesproduced in bacteria)

� Prb

Declerck.2

binding to relevant Fc gamma receptors, the func-

tion of the “Fab” portion (including receptor func-tion and ligand neutralization), as well as the Fc-

mediated actions such as complement activation,

ADCC, and CDC functions.2

Because murine-derived mAbs may be potentially

immunogenic in humans and may lack necessary

Fc-mediated actions, many mAbs are fully human(eg, panitumumab) or have been engineered

through recombinant DNA technology to be chi-

meric (ie, having mouse and human portions; eg,rituximab) or humanized (eg, trastuzumab).2 Differ-

ences in immunogenicity between biosimilar mAbs

and the reference mAbs is important to consider, asthese differences could lead to loss of efficacy,

serious AEs, or death associated with the biosimilar

mAb.7 The higher level of complexity associatedwith developing and evaluating biosimilar mAbs

poses several challenges that need to be overcome

ilar mAbs to Market

Anticipated Impact

igher costs of development resulting in muchmaller difference in price between the biosimilarnd reference biologic productmaller perceived benefit of substituting a first-eneration biosimilar product versus a second-eneration productmaller perceived benefit of substituting a first-eneration biosimilar product

otential for further lowering production costs foreference biologic product versus producing theiosimilar product

Page 9: Developing Oncology Biosimilars: An Essential Approach for the Future

Developing oncology biosimilars S13

if biosimilar mAbs are to gain widespread use in

clinical practice (Table 4).2

CHALLENGES ASSOCIATED WITHINTEGRATING BIOSIMILARS INTO CLINICALPRACTICE

Key challenges associated with integrating biosi-

milars into clinical practice may include multipleFDA-approved indications for the reference biologic

products, off-label uses of the reference biologic

products, economic and uptake issues, substitutionpractices, and the need for HCP education (Table 5).

Many of these challenges relate to the way biosimi-

larity will be defined and demonstrated. For exam-ple, a key question is how similar is “highly similar,”which means how much evidence must be provided

to gain regulatory approval for biosimilars; thisquestion can also be applied to another important

issue, the interchangeability of the biosimilar and the

reference biologic product from a prescribing stand-point.17 According to the 351(k)(4) of the PHS Act,

biosimilars may be considered “interchangeable” if

an applicant provides sufficient information to dem-onstrate biosimilarity and that the biosimilar can be

expected to produce the same clinical result as the

reference biologic product in any given patient, and,if the biosimilar is administered more than once to

an individual, the risk in terms of safety or dimin-

ished efficacy of alternating or switching betweenthe use of the biosimilar and the reference biologic

product is not greater than the risk of using the

reference biologic product without such alternationor switch. Because such similarity would be difficult

to clinically demonstrate, alternative definitions have

been suggested, such as producing the same ther-apeutic effect within a reasonable degree of statis-

tical assurance. From a practical standpoint, data

from bioanalytical, animal, and clinical studies maybe used to demonstrate biosimilarity with a refer-

ence biologic product.1 Appropriate guidance from

the EMA or the FDA regarding the design andrequirements for biosimilar bioanalytical studies

has yet to be provided.7

Safety and Pharmacovigilance Issues

Pharmacovigilance refers to scientific and data

collection activities related to the detection,

assessment, understanding, prevention, and com-munication of potential safety concerns with drug

products.24,39,40 The goal of pharmacovigilance is

to promptly identify and evaluate safety signals sothat risks can be appropriately managed.39 Phar-

macovigilance programs for biosimilars should

include strategies for AE tracking and tracing topromote responses such as recalls and/or alerts.

This traceability requires the adoption of a rational

international nomenclature system for biosimilars,which has yet to be fully elucidated.39 Such bio-

similar nomenclature may include a unique non-

proprietary name, the Healthcare Common Pro-cedure Coding System/NDC number and the lot

number for each biosimilar; this is needed to

ensure that any potential AE associated with aparticular biosimilar (ie, molecule-specific) is not

erroneously attributed to an entire class of bio-

logics.39 For example, in the EU, use of the Inter-national Nonproprietary Name (INN), which iden-

tifies active pharmaceutical substances and is

crucial for drug substitutions and postmarketingsurveillance reports, as the reference biologic

product may result in safety issues for patients

(eg, AE reporting).13 In addition, pharmacovigi-lance systems must have the flexibility and real-

time data collection capabilities to identify unique

and/or unexpected AEs associated with the bio-similar versus the reference biologic product (eg,

AEs potentially resulting from immunogenicity).39

Ultimately, education of HCPs on pharmacovigi-lance procedures will be required, as they will

bear much of the responsibility for accurately

identifying, recording, and reporting an AE asso-ciated with a biosimilar.1,39

Although it is not possible to predict how

differences in manufacturing or purification of agiven biosimilar will ultimately impact its efficacy

and safety, some insight can be gleaned from the

safety experience with biosimilars in Europe.9,39

For example, most EU countries, in cooperation

with WHO resources, have developed post-

marketing pharmacovigilance programs enablingHCPs, and in some cases patients, to report

any unanticipated AEs and/or lack of efficacy

for a particular biosimilar.30 This type of riskassessment may be particularly useful for detect-

ing rare AEs only observed in large numbers of

patients.30

In Europe, exposure to biosimilars has been

steadily accumulating: for example, as of 2013,

exposure (Binocrit, Sandoz International GmbH,BU Biopharmaceuticals, Holzkirchen, Germany)

was 216,000 patient-years with more than 5,000

patients studied, and exposure to one biosimilarfilgrastim (Zarzio, Sandoz International GmbH,

Kundl, Austria) was 3.5 million patient-days.3 To

date, there have been no relevant efficacy or safetyconcerns with the use of biosimilar versions of

filgrastim in Europe.16 The use of biosimilar fil-

grastims for the prophylaxis of febrile neutropeniais now addressed in the European Organization for

Research and Treatment of Cancer (EORTC) guide-

lines; however, their use is regarded as a change intherapy owing to the possibility of differences

Page 10: Developing Oncology Biosimilars: An Essential Approach for the Future

Table 5. Challenges Associated With Incorporating Biosimilars Into Oncology Practices in theUnited States

Topic Challenges

Clinical trials for biosimilars � Although the amount of data needed to approve biosimilardrugs will be less than that needed for a new biologic drug, itwill be greater than that needed for a small-molecule genericdrug; it is expected that clinical trials will be required.However, overall clinician and patient interest in enrolling insuch trials may be low

Economic considerations ofbiosimilars

� Cost-savings with biosimilars will be less than that seen forgeneric small-molecule drugs, because manufacturing anddevelopment costs will be higher; it is unclear how payors willdeal with coverage and reimbursement issues related tobiosimilars; competing cost reductions by the referencebiologic product manufacturer also could impact the overallsavings realized by substituting biosimilars

Safety and pharmacovigilance � Because biosimilars cannot be considered completely identicalto their reference biologic product, they cannot be fullycharacterized from a safety perspective and rigorouspharmacovigilance procedures are needed

Tracking of biosimilars in clinicalpractice

� Tracking the source of biologic drugs (ie, biosimilar vs thereference biologic product) may be challenging, particularlyin community settings, which lack the tracing infrastructureused at larger institutions (ie, which drug was used in whichpatient); effective tracking also requires the reference biologicproduct and the biosimilar to be named differently withdifferent drug codes

Substitution of biosimilars � The FDA may determine that a biosimilar is interchangeablewith a given reference biologic product, however, substitutionpractices vary based on state laws, and these practices will bedifferent from those used for small-molecule generic drugs

Extrapolation of data for other approvedindications or off-label use

� Based on data submitted for 1 indication, the FDA willdetermine whether the use of biosimilars can be extrapolatedto all approved indications of a reference biologic product; itis unclear whether clinicians, payors, and other stakeholderswill consider such data sufficient for extrapolation to otherindications or off-label use

Patient and provider education � The manufacturing and development of biologic drugs andbiosimilars is complex, and physicians need to be aware of thedifferences between biosimilars and generic drugs; educationon the use of biosimilars is needed

Zelenetz et al,1 Schellekens,4 Niederweiser et al,15 Blackstone et al.27

J. AbrahamS14

between the biosimilar and the reference biologic

product. As such, a biosimilar filgrastim cannot be

exchanged without physician and patientknowledge.16

Immunogenicity

The overall immunogenicity of biosimilars can be

impacted by the manufacturing and purificationprocess.3,16 Given that slight differences in biologics

may occur as a result of different manufacturing

processes, it is important to evaluate the immunoge-

nicity of a biosimilar versus the reference biologicproduct.2,10 As shown in Figure 7, subtle variations in

manufacturing and/or storage conditions could have

an impact on the immunogenicity of biologics.41

Immunogenicity studies should include an assess-

ment of the presence of antidrug antibodies (ADAs)

and a determination of whether these ADAs act asneutralizing antibodies that effectively reduce or even

Page 11: Developing Oncology Biosimilars: An Essential Approach for the Future

Figure 7. Immunogenicity in patients treated with dif-ferent interferon alpha preparations. Abbreviations: HSA,human serum albumin; IFN, interferon. Refer to the webversion of this article at seminoncol.org for color figure.Reprinted by permission from Macmillan Publishers Ltd:[Seminars in Oncology], Schellekens H. Bioequivalenceand the immunogenicity of biopharmaceuticals. Nat RevDrug Discov. 2002;1:457–62,41 copyright 2002.

Developing oncology biosimilars S15

eliminate activity.7 Specifically, the presence of ADAs

can impact the clinical response to a biologic by

increasing or decreasing drug clearance, neutralizingthe pharmacologic action of the drug and potentially

an endogenous cross-reactive counterpart (which

may lead to a potential deficiency syndrome), orcausing an immune response or hypersensitivity

reaction.7 Development of immunogenicity assays is

challenging because there is more than one drugproduct being assessed (ie, the biosimilar and one or

more reference biologic products).7 Assessment of

immunogenicity is also hampered by a lack of stand-ardization among different laboratories (eg, types of

assays used and methods used to report the data).24

Pharmacokinetic and pharmacodynamic data, aswell as clinical AE profile data, also must be consid-

ered when interpreting immunogenicity.7 Both the

FDA and the EMA require that results of comparativestudies demonstrate that the biosimilar is no more

immunogenic than the reference biologic product.7

Despite these requirements and regardless of theresults of immunogenicity testing, the postmarketing

pharmacovigilance plan, such as the plan developed

in the EU, will be key to the safe use of biosimilars,especially biosimilar mAbs. This is especially impor-

tant because there currently are no means of reliably

predicting immunogenicity in humans, and thereforesuch assessments must be based on risk.2,38

Of note, in 2002 and 2003, European patients with

chronic renal failure who received rhEPO developedan erythropoietin-resistant, transfusion-dependent

severe anemia caused by antibody-mediated pure red

cell aplasia.39,42,43 Most of these cases ultimately wereattributed to a specific preparation of erythropoietin

made by a single manufacturer, and among the

possible causes identified were differences in manu-facturing, formulation, and the primary container

used.38,44,45 Although these experiences were based

on a slight change in the manufacturing process andnot the use of a biosimilar, they highlight the need for

immunogenicity testing when assessing biosimilarity,

as well as the importance of having rigorous pharma-covigilance procedures in place to detect and track

unanticipated AEs.24

Provider and Payor Issues

Ensuring that providers have confidence in the

safety and efficacy of biosimilars is paramount.Physicians, pharmacists, and payors are likely to

have the most impact on the use and assimilation

of biosimilars into clinical practice, and they must beconfident in the data provided to regulatory agencies

for approval (including demonstrations of biosimi-

larity and interchangeability with the reference bio-logic product regarding safety and efficacy), as well

as the resulting regulatory agency guidance.1 It is

especially important that HCPs understand the com-plexities involved in producing biologic drugs and

the potential clinical implications of using biosimi-lars that are considered similar, but not exactly

identical, to the reference biologic product.1,8 As

such, an in-depth understanding of biosimilars as adrug class will be required for physicians, pharma-

cists, and those responsible for reimbursement of

treatment costs.16 Evidence from the NCCN suggestthat significant knowledge gaps currently exist.1

Nomenclature Issues

Generic drugs, by definition, have the same non-

proprietary name as the original reference drug.1

Because biosimilars are not identical to referencebiologics, one possible naming convention is that

biosimilars should be assigned a related, but unique

and distinguishable nonproprietary name. Thisapproach may facilitate postmarketing safety mon-

itoring and AE data collection by ensuring that

specific events can be correctly attributed to thebiosimilar or the reference biologic.1 For example,

Page 12: Developing Oncology Biosimilars: An Essential Approach for the Future

J. AbrahamS16

for naming biosimilars in Japan, both the brand name

and the nonproprietary name of the reference bio-logic product are used, with the nonproprietary

name followed by “kozoku-1,” which means follow-

on biologic-1 or biosimilar-1 (eg, Avastin bevacizu-mab kozoku-1).45

Since biosimilars cannot be considered identical

to reference biologics, the United States AdoptedName Council (USANC) has stated that biosimilar

manufacturers should not assume biosimilars will

have the same nonproprietary names as the refer-ence biologics.1 Guidance from the regulatory agen-

cies is needed to determine whether a biosimilar and

its reference biologic will have the same nonpropri-etary name.1 One disadvantage to the unique naming

of biosimilars is the potential for confusion among

HCPs and patients regarding comparability with thereference biologic products; even if biosimilarity has

been adequately proven, different names may bias

HCPs and/or patients into thinking that safety andefficacy is not equivalent.1

Regulations Regarding Biosimilar Substitution

The process (ie, policies and regulations) for

substituting the reference biologic for a biosimilarmay be difficult for HCPs to navigate. Although the

BPCI Act allows the FDA to make an interchange-

ability determination for biosimilars, substitutionpractices are governed by state laws.1 Current state

laws regarding automatic drug substitution (ie, the

substitution of a generic for the original referencebiologic product unless the prescriber specifies

otherwise) were developed and enacted before the

emergence of biosimilars.1 As such, individual statesmay or may not take into account FDA-directed

interchangeability determinations when developing

new laws and regulations governing the use ofbiosimilars, which could lead to further confusion

among HCPs.1

THE BIOSIMILAR EXPERIENCE IN COUNTRIESOTHER THAN THE UNITED STATES

The integration of biosimilars has already begun incountries other than the United States, putting the

US government, biotechnology, and pharmaceutical

industries in a position to learn from these experi-ences and develop effective manufacturing and

approval processes that facilitate the safe integration

of biosimilars into clinical practice. Although indi-vidual requirements, definitions, and scope of guid-

ance may differ, most regulatory authorities around

the world appear to adhere to several key principlesregarding the characterization of biosimilars17:

The regulatory approach to biosimilars should bedifferent from that used for generic drugs, and

similarity in terms of quality, efficacy, and safety

must be proven for the biosimilar and the refer-ence biologic product

As a prerequisite for reducing the amount of

nonclinical and clinical data submitted for appro-

val, a stepwise approach is needed to ensureadequate similarity in quality between the biosi-

milar product and the reference biologic product

Different classes of biologics may necessitate

distinct biosimilarity assessment requirements

Close attention to pharmacovigilance is neededfor biosimilars

The EU was an early adopter of biosimilars and

provides examples of what the regulatory process

will likely entail for the United States. In addition, anumber of emerging markets have some experience

with biosimilars. In Brazil, a two-pathway system

was developed in 2010 to help foster the use ofbiosimilars.46 The first of these pathways, termed the

“individual pathway,” was developed in response to

high expenditures for pegylated interferons for thetreatment of hepatitis C. In this pathway, the devel-

opment process, dossier, quality issues, and require-

ments for clinical studies are reduced, butindications cannot be extrapolated.46 The second

pathway, termed the “comparability pathway,”allows extrapolation to other indications; however,the pathway is more rigorous and requires compa-

rative phase I, II, and III clinical trials with the

reference biologic product.46 While the adoption ofthese pathways may help drive the integration and

utilization of biosimilars, it is not yet known how

these regulatory pathways might be impacted withdevelopment of more complex biosimilars such as

biosimilar mAbs.46

China and India represent two very large andemerging markets for biosimilars; however, there

may be some unique challenges as Western bio-

similar manufacturers enter these markets and viceversa.18 In China, domestically produced biosimi-

lars have been available for use for more than 20

years, and there is no specific regulatory processfor approval, with the State Food and Drug Admin-

istration overseeing a similar regulatory process for

all pharmaceuticals.18 In India, there has been arobust acceptance of biosimilars, with some 50

biopharmaceuticals on the market, of which

greater than half are biosimilars; nonetheless, someconcern has been raised over the standards of the

Indian regulatory authorities in terms of the purity

of its biosimilars and control over postmanufactur-ing storage and distribution conditions.18,47 Indian

biosimilar manufacturers will need to adhere to

EMA guidelines as they begin to enter the globalbiosimilars market.47

Page 13: Developing Oncology Biosimilars: An Essential Approach for the Future

Developing oncology biosimilars S17

EXAMINING THE CURRENT STATE OF AFFAIRS

The NCCN Biosimilars Work Group

In 2011, the NCCN formed a Biosimilars Work

Group consisting of a multidisciplinary group ofproviders (physicians, pharmacists, and nurses), man-

ufacturers, patients, payors, and government offi-

cials.1 The purpose of the work group was toadvise oncology practitioners and other key stake-

holders on the major challenges ahead and provide

recommendations for developing and using biosimi-lars in oncology.1 The key consensus statements and

recommendations of the NCCN Biosimilars Work

Group are summarized in Tables 6 and 7.1 Briefly,with regard to recommendations of the work group,

it is expected that clinical trials incorporating rele-

vant and sensitive end points will be needed to

Table 6. Biosimilars in Oncology: Consensus Statem

Importance, access, andaffordability

● The overall goal ofto biologic medicatherapies for cance

● The NCCN Work Goncology care andpathway (character

Approval pathway shoulddemonstrate similarity

● At the time of bioshigh similarity to thpharmacodynamic

● Efficacy and safety ono clinically meaninreference biologic p

Standardization with referencebiologic product

● The NCCN Work Gelements of a biosibiologic product (fomedication errors).should be the sameAdditionally, Risk Ethe associated provbetween biosimilar

Tracking product use ● The ability to trackroutine clinical usebatch was seen assafety of these med

Need for educating patients andHCPs

● Patients and HCPs rof biosimilars. Patiereceive

● In the context of mhelp avoid medicatthey receive

Zelenetz et al.1

demonstrate similarity between the biosimilars and

the reference biologic product.1 It was also agreedthat guidance is needed from the FDA on key issues

such as specifically defining what is meant by a “highdegree of similarity” and “no clinically meaningfuldifferences in efficacy and safety.”1 It is also expected

that NCCN guideline panels will need to evaluate

recommendations about the use of biosimilars rela-tive to specific diseases to provide guidance for

pharmacy and therapeutics committees and other

practitioners in community practice. It also wasrecognized that educational activities are required

to inform HCPs, including physicians, nurses, phar-

macists, physician assistants, and nurse practitionersof the development, evaluation, and pharmacovigi-

lance efforts that will be necessary to make biosimi-

lars viable options for the treatment of malignances.1

ents From the NCCN Biosimilars Work Group

biosimilars is to increase affordability and accesstions for patients, which are often importantr careroup believes biosimilars are important tois supportive of defining a biosimilars approvalized in the BPCI Act of 2009)

imilar approval, a biosimilar must have showne reference product in quality attributes andand pharmacokinetic parametersf the biosimilar product must be comparable (ie,gful difference in safety and efficacy) to theroduct

roup agrees with current regulations thatmilar drug product should follow the referencer the purpose of consistency of practice avoidingFor example, the dosing for a biosimilar agentas for the reference biologic product.

valuation and Mitigation Strategies (REMS) andider workflow process should be standardizedand reference biologic products

a patient’s receipt of a biosimilar product duringdown to the level of a specific manufacturer anda critical element of assessing and ensuring theications

equire education to increase their understandingnts also should know all the medicines they

ultisourced biologics being available, this mayion errors if patients know the exact drug product

Page 14: Developing Oncology Biosimilars: An Essential Approach for the Future

Table 7. Considerations and Recommendations Regarding Biosimilars in Oncology: The NCCNBiosimilars Work Group

Considerations Recommendations

Use of appropriate endpoints

� End points studied must be sensitive enough to show differences, if any,between biosimilar and the reference biologic product; end points suchas overall response, overall survival, and/or progression-free survival willbe most helpful for HCPs

Consistency andtransparency

� A consistent and transparent approach is recommended for the FDAprocess of determining biosimilarity, including definitions of highly similarquality attributes and “no meaningful differences in efficacy or safety”;required scientific data should be based on known safety and efficacyprofile of the reference biologic product

Use in NCCN Guidelines � NCCN Guideline Panels should evaluate biosimilars and their potentialrole in treatment and provide recommendations for their use;recommendations against biosimilars are not anticipated, but informationwill be helpful to provide clarity for key stakeholders (ie, clinicians,patients, payors)

Pharmacy and Therapeutics(P&T) Committees

� Institutional P&T committees should evaluate biosimilars for use in theirspecific populations and this approach should be different from that ofgeneric small-molecule drugs; as with other drugs, for those practitionersnot under P&T guidance, consideration and review of individualbiosimilars is recommended before routine use is implemented

HCP and policymakereducation

� Education about basic scientific principles of biologic drugs and biosimilars,as well as their manufacturing processes and associated pharmacovigilanceprocedures, should be disseminated to HCPs (ie, physicians, pharmacists,nurses), in addition to legislators and policy makers

Safety, tracking, andnomenclature

� Guidance is needed from the FDA about naming of biosimilars (eg, willthey have unique nonproprietary names)

Zelenetz et al.1

J. AbrahamS18

WHO and FDA Guidance

In 2010, the WHO provided guidance for evaluat-

ing “similar biotherapeutic products” or SBPs.35 Thisguidance recognizes the differences betweengeneric drugs and biosimilars and the processes

required to conclude their therapeutic equiva-

lence.35 Some of the key principles of the WHOguidance regarding the licensing of biosimilars are

listed in Table 8.35 The BPCI Act, by comparison,

only serves to outline broad concepts related to USbiosimilar approval policy, and it was recognized

that additional information from the FDA was

needed.48

A significant step toward integrating biosimilars

into clinical practice was made by the FDA with the

release in 2012 of draft guidance for the develop-ment and approval of biosimilars.19 Further draft

guidance on biosimilars was provided by the FDA in

2012, but some important questions remain.48 In2013, Li and Hoffman reviewed the draft FDA

guidance from the point of view of practicing

oncologists.48 The main purpose of the first FDAdocument, Quality Considerations in Demonstrating

Biosimilarity to a Reference Protein Product is to provide

information on determining biosimilarity to thereference biologic product.48 The document specif-

ically summarizes the FDA’s position on demonstrat-

ing biosimilarity to a reference biologic product thatis listed under Section 351(a) of the PHS Act, for the

purposes of submitting a marketing application

under Section 351(k) of the PHS Act. The documentalso provides the definition of biosimilarity included

in Table 1.34 The document concludes that the

demonstration of biosimilarity with a reference bio-logic product involves “the robust characterization of theproposed biosimilar product, including comparative physico-chemical and functional studies.”34

The second document, Scientific Considerations inDemonstrating Biosimilarity to a Reference Product, pro-vides information on the types of evidence requiredto demonstrate biosimilarity.19,48 The proposed evi-

dence to be provided includes structural and func-

tional analyses comparing the biosimilar with thereference biologic product, as well as in vivo animal

studies comparing toxicity, pharmacokinetic, phar-

macodynamic, and potentially immunogenicityparameters between the products.19,48 Also of

Page 15: Developing Oncology Biosimilars: An Essential Approach for the Future

Table 8. WHO Guidance on Biosimilars: Key Principles Regarding the Licensing of Similar BiologicProducts

● The development of a SBP involves stepwise comparability exercise(s) starting with comparison of thequality characteristics of the SBP and RBP. Demonstration of similarity of a SBP to a RBP in terms ofquality is a prerequisite for the reduction of the nonclinical and clinical data set required for licensure.After each step of the comparability exercise, the decision to proceed further with the development ofthe SBP should be evaluated

● The basis for licensing a product as a SBP depends on its demonstrated similarity to a suitable RBP inquality, nonclinical, and clinical parameters. The decision to license a product as a SBP should bebased on evaluation of the whole data package for each of these parameters

● If relevant differences are found in the quality, nonclinical or clinical studies, the product will not likelyqualify as a SBP and a more extensive nonclinical and clinical data set will likely be required to supportits application for licensure. Such a product should not qualify as a SBP as defined in this guideline

● If comparability exercises and/or studies with the RBP are not performed throughout the developmentprocess as outlined in this guidance document, the final product should not be referred to as a SBP

● SBPs are not “generic medicines” and many characteristics associated with the authorization processgenerally do not apply

● SBPs, like other biotherapeutic products, require effective regulatory oversight for the management ofpotential risks and to maximize benefits

Abbreviations: RBP, reference biologic product; SBP, similar biologic product.WHO.35

Developing oncology biosimilars S19

importance in this draft guidance is the requirementfor human clinical studies aimed at showing “noclinically meaningful differences between the [biosimilar]product and the reference biologic product in terms of safety,purity, and potency.”19,48 The document suggests a

need for clinical pharmacokinetic/pharmacodynamic

data, clinical efficacy and safety data, and clinicalimmunogenicity data, as well as postmarketing sur-

veillance measures for biosimilars.19,48 The docu-

ment also notes that specific clinical/preclinicaldata required will differ depending on the type of

product, and that a “totality of the evidence”approach will be used to consider biosimilars.19,48

With such an approach, all data submitted by the

sponsor, with regard to structural and functional

characterization, nonclinical evaluation, humanpharmacokinetic and pharmacodynamic data, clini-

cal immunogenicity data, and clinical safety and

efficacy data will be evaluated.19,34

Some of the key limitations of this draft FDA

guidance include a lack of advice on what types of

clinical trials will be required for which products,although each biologic will likely be assessed on a

case by case basis, with more complex biologic

products (eg, mAbs) requiring additional evalua-tion compared with simpler products.2,19,48

Current automatic substitution policies are notapplicable to biosimilars; further guidance is

needed in this area. There also may be a need

for additional explicit guidance to individualstates on the substitution of biosimilars without

notification of the prescriber or patient.48 In

addition, questions may arise if a product isdemonstrated to be a biosimilar for only one of

several indications held by the reference biologic

product. Whether or not clinical trials for otherindications will be mandated by the FDA is unclear

(Table 5).48 Some of the key points in the FDA

guidance from 2012 are summarized in Table 9.7

In March 2013, the FDA issued further guidance

titled Formal Meetings Between the FDA and Biosimi-lar Biological Product Sponsors or Applicants. Thepurpose of this document is to provide nonbind-

ing recommendations to industry regarding formal

meetings between the FDA and biosimilar spon-sors or applicants.49

The NCCN Survey

In 2011, the NCCN published the results of a

survey on HCP knowledge of biosimilars. This surveywas conducted at the NCCN 16th Annual

Page 16: Developing Oncology Biosimilars: An Essential Approach for the Future

Table 9. FDA Draft Guidance on Biosimilars: Key Points

● FDA will use a risk-based, totality of evidence approach when evaluating data to determinebiosimilarity

● Sponsors should develop a stepwise approach to demonstrating biosimilarity, including structural andfunctional characterization of the biosimilar and the reference biologic product (eg, post-translationalmodifications)

● A full nonclinical program may not be needed if extensive structural and functional similarity isdemonstrated in comparative studies and animal toxicology studies

● Comparative clinical studies must demonstrate purity, potency and safety though an evaluation ofimmunogenicity, pharmacokinetics, and pharmacodynamics in a condition for which the referencebiologic product is approved

Cai et al.7

Figure 8. NCCN Survey: Biosimilars – Response to:“Please rate your overall familiarity with developmentsfor biosimilars, including recent legislation that providesan approval pathway for nonreference (eg, ‘generic’)manufacturers to introduce copies of biologics throughan abbreviated review process.” Refer to the web versionof this article at seminoncol.org for color figure. ZelenetzAD, Ahmed I, Braud EL, et al. NCCN Biosimilars WhitePaper: regulatory, scientific, and patient safety perspec-tives. J Natl Compr Canc Netw. 2011;9(Suppl 4):S1–22,1

reprinted with permission from JNCCN—Journal of theNational Comprehensive Cancer Network.

J. AbrahamS20

Conference (March 2011), and 277 participants

responded including physicians (47%), nurses

(26%), and pharmacists (14%); a smaller percentageof other clinicians and nonclinicians also responded.1

Respondents were asked about their overall familiarity

with developments around biosimilars, includingawareness of legislation providing for abbreviated

approval.1 The results indicated that 55% of respond-

ents were either not at all familiar or only slightlyfamiliar with the developments around biosimilars

(Figure 8).1

The respondents’ overall interest in prescribing,dispensing, and administering biosimilars, once FDA

approved for use, also was assessed. The results

indicated a high level of interest (62% rating theirinterest as high or moderately high) (Figure 9).1

Notably, just over a quarter of the respondents said

they need more information about biosimilars beforemaking their decision.1 Respondents were asked

to evaluate the importance of specific types of

information when making decisions about biosimi-lars.1 Results for this question showed that, whereas

studies comparing clinical end points in safety and

efficacy appeared most important (with 74% ratingthis type of information as very important), 86%

of respondents indicated that all the types of infor-

mation identified in the survey were at least some-what or very important (Figure 10).1

Additional data from the survey also highlighted

those respondents who were familiar with biosimilardevelopments had defined opinions on whether they

would immediately use or not use biosimilars once

they become available. Most respondents who hadonly somewhat or slight familiarity with biosimilars

stated they would require further review and discus-

sion before making a decision to use biosimilars.1 TheNCCN noted that because a “convenience sample” (ie,

surveying a nonspecific group) was used, the respond-

ents may not be representative of US oncology

practitioners. This may be a reason why their overallknowledge of biosimilars was fairly low.1 Despite the

limitations, the overall conclusions from this survey

indicate that while there was a high level of interest inusing biosimilars, awareness of biosimilars is subopti-

mal, and a need exists for greater patient and provider

education about these emerging products.1

Page 17: Developing Oncology Biosimilars: An Essential Approach for the Future

Figure 9. NCCN Survey: Biosimilars – Response to: “Onceapproved by the FDA, what is your interest in prescribing,dispensing, or administering biosimilars in your practice set-ting?” Refer to the web version of this article at seminoncol.org for color figure. Zelenetz AD, Ahmed I, Braud EL, et al.NCCN Biosimilars White Paper: regulatory, scientific, andpatient safety perspectives. J Natl Compr Canc Netw. 2011;9(Suppl 4):S1–22,1 reprinted with permission from JNCCN—Journal of the National Comprehensive Cancer Network.

Developing oncology biosimilars S21

BIOSIMILARS: COMPETING INTERESTS, RISKS,AND BENEFITS

The development and implementation of biosimi-lars is of interest to a wide range of stakeholders,

including the patients who will ultimately use them,

HCPs who prescribe them, manufacturers of thereference biologic products and biosimilars, as well

as payors and government regulatory agencies.1 The

successful integration of biosimilars into the

Figure 10. NCCN Survey: Biosimilars – Response to: “As more infothe following types of information in helping you decide to use bioseminoncol.org for color figure. Zelenetz AD, Ahmed I, Braud EL, etpatient safety perspectives. J Natl Compr Canc Netw. 2011;9(Supplthe National Comprehensive Cancer Network.

pharmaceutical marketplace will require a consensus

among all the stakeholders involved.1 The benefits ofinnovation, expanded drug access, and affordability of

medications must be balanced with the need for

sufficient safety and efficacy data, as illustrated inFigure 11.1 For example, from an economic stand-

point, an extensive verification process may impact

production costs, resulting in less pharmacoeconomicbenefit versus the reference biologic product. From

the patient and HCP perspective, a more extensive

verification process will provide more comprehensivesafety and efficacy data versus the reference biologic

product, which may allow greater patient/provider

acceptance and integration into practice. The sameset of examples also can be applied from the insurers’and payors’ perspective, and thus it remains to be

seen how these factors will come together to impactthe ultimate success of biosimilars.

SUMMARY AND CONCLUSIONS

While biologics have helped revolutionize the

treatment of cancer, they are expensive, accountingfor more than half of the healthcare expenditures

in oncology. The Patient Protection and Affordable

Care Act, signed into law by President Obama onMarch 23, 2010, has amended the PHS Act and

provided for an abbreviated approval pathway for

biologics that are demonstrated to be “biosimilar” or“interchangeable” with an FDA-licensed reference

biologic product. This has helped pave the way for

the eventual emergence of biosimilars in the UnitedStates.19 The development and implementation of

biosimilars has the potential to create a truly com-

petitive and innovative environment in which the

rmation on biosimilars becomes available, how important aresimilar products?” Refer to the web version of this article atal. NCCN Biosimilars White Paper: regulatory, scientific, and4):S1–22,1 reprinted with permission from JNCCN—Journal of

Page 18: Developing Oncology Biosimilars: An Essential Approach for the Future

Figure 11. Biosimilars: risks and benefits in the development and approval process. Zelenetz et al.1

J. AbrahamS22

cost of biologics may be reduced, increasing their

overall affordability and accessibility for patients. It isclear that access to therapy is an important predictor

of outcome and that the availability of lower cost

biosimilar treatments could make treatments an optionfor more patients earlier in the course of disease.

The process involved in developing biosimilars is,

however, markedly different from that for producinggeneric versions of small-molecule drugs, which has

important implications for manufacturing, approval,

regulatory, and substitution processes. In addition,several key oncology drugs are mAbs, which have

many levels of molecular complexity that make them

especially challenging to produce as biosimilars.There also are several regulatory issues associated

with the development and implementation of bio-

similar drugs that remain unresolved. Overall,because they involve a more complex development

process, the level to which they will reduce costs

over the reference biologic products is likely to beless than that of generics. How biosimilar products

will be received by patients and providers remains to

be seen. Because they can be considered “highlysimilar” but not exactly identical to the reference

biologic product, new policies regarding the nomen-

clature of biosimilars, as well as extensive pharma-covigilance procedures to quickly identify any

important differences in efficacy and/or safety from

the reference biologic product, are needed.Results from a recently published NCCN survey

suggest that, despite a high interest in the potential

benefits of using biosimilar drugs, there is a need forgreater education on biosimilars for oncologists,

nurses, pharmacists, and other HCPs. It will be

essential that all key stakeholders, including physi-cians, pharmacists, and payors, possess an in-depth

understanding of biosimilars so that the potential for

improving access and outcomes in oncology can berealized. Important additional guidance from the

FDA regarding biosimilars is also needed and

expected and this will be critical to the furtherdevelopment of biosimilars in the United States.

REFERENCES1. Zelenetz AD, Ahmed I, Braud EL, et al. NCCN Bio-

similars White Paper: regulatory, scientific, and patient

safety perspectives. J Natl Compr Canc Netw. 2011;9

(Suppl 4):S1–22.

2. Declerck PJ. Biosimilar monoclonal antibodies: a

science-based regulatory challenge. Expert Opin Biol

Ther. 2013;13:153–6.

3. Generics and Biosimilar Initiative. Biosimilars in oncol-

ogy: current and future perspectives. gabi-journal.net/

biosimilars-in-oncology-current-and-future-perspectives.

html. Accessed July 24, 2013.

4. Schellekens H. Biosimilar therapeutics–what do we

need to consider? NDT Plus. 2009;2(Suppl 1):i27–36.

5. United States House of Representatives. United States

Public Health Services Act. www.house.gov/legcoun/

Comps/PHSA_CMD.pdf. Published March 13, 2013.

Accessed July 19, 2013.

6. Generics and Biosimilar Initiative. FDA definitions of

generics and biosimilars. http://gabionline.net/Biosimi

lars/General/FDA-definitions-of-generics-and-biosimilars.

Accessed July 19, 2013.

7. Cai XY, Thomas J, Cullen C, Gouty D. Challenges of

developing and validating immunogenicity assays to

support comparability studies for biosimilar drug

development. Bioanalysis. 2012;4:2169–77.

8. Lee JF, Litten JB, Grampp G. Comparability and

biosimilarity: considerations for the healthcare pro-

vider. Curr Med Res Opin. 2012;28:1053–8.

9. Kuhlmann M, Covic A. The protein science of biosimi-

lars. Nephrol Dial Transplant. 2006;21(Suppl 5):v4–8.

Page 19: Developing Oncology Biosimilars: An Essential Approach for the Future

Developing oncology biosimilars S23

10. Kanter J, Feldman R. Understanding and incentivizing

biosimilars. Hastings Law J. 2012;64:57–83.

11. Verpoort K, Mohler TM. A non-interventional study of

biosimilar granulocyte colony-stimulating factor as

prophylaxis for chemotherapy-induced neutropenia

in a community oncology centre. Ther Adv Med

Oncol. 2012;4:289–93.

12. Ward E, Halpern M, Schrag N, et al. Association of

insurance with cancer care utilization and outcomes.

CA Cancer J Clin. 2008;58:9–31.

13. Jelkmann W. Biosimilar epoetins and other "follow-on"

biologics: update on the European experiences. Am J

Hematol. 2010;85:771–80.

14. McCamish M, Woollett G. The continuum of compa-

rability extends to biosimilarity: how much is enough

and what clinical data are necessary? Clin Pharmacol

Ther. 2013;93:315–7.

15. Niederwieser D, Schmitz S. Biosimilar agents in oncol-

ogy/haematology: from approval to practice. Eur J

Haematol. 2011;86:277–88.

16. Hus I. Follow-on biologics in oncology—the need for

global and local regulations. Wspolczesna Onkol.

2012;16:461–6.

17. Wang J, Chow S-C. On the regulatory approval path-

way of biosimilar products. Pharmaceuticals. 2012;5:

353–68.

18. Generics and Biosimilar Initiative. Hurdles to biosimi-

lars in Asia. http://gabionline.net/Biosimilars/General/

Hurdles-to-biosimilars-in-Asia. Accessed July 19, 2013.

19. United States Food and Drug Administration. Guidance

for industry: quality considerations in demonstrating

biosimilarity to a reference protein product. http://

www.fda.gov/downloads/Drugs/GuidanceCompliance

RegulatoryInformatin/Guidances/UCM291134.pdf. Ac-

cessed July 19, 2013.

20. Fedewa SA, Lerro C, Chase D, Ward EM. Insurance

status and racial differences in uterine cancer survival:

a study of patients in the National Cancer Database.

Gynecol Oncol. 2011;122:63–8.

21. Kozlowski S, Woodcock J, Midthun K, Sherman RB.

Developing the nation’s biosimilars program. N Engl J

Med. 2011;365:385–8.

22. Federal Trade Commission. Emerging health care

issues: follow-on biologic drug competition. Washing-

ton, DC: Federal Trade Commission; 2009. http://

www.ftc.gov/os/2009/06/P083901biologicsreport.pdf.

Accessed July 19, 2013.

23. Mellstedt H, Niederwieser D, Ludwig H. The challenge

of biosimilars. Ann Oncol. 2008;19:411–9.

24. Locatelli F, Roger S. Comparative testing and pharma-

covigilance of biosimilars. Nephrol Dial Transplant.

2006;21(Suppl 5):v13–6.

25. Generics and Biosimilar Initiative. US$54 billion

worth of biosimilar patents expiring before 2020.

http://www.gabionline.net/Biosimilars/Research/US-

54-billion-worth-of-biosimilar-patents-expiring-before-

2020. Accessed July 19, 2013.

26. Doloresco F, Fominaya C, Schumock GT, et al. Projec-

ting future drug expenditures: 2011. Am J Health Syst

Pharm. 2011;68:921–32.

27. Blackstone EA, Fuhr JP Jr. Innovation and competition:

will biosimilars succeed? Biotechnol Healthc. 2012;9:24–7.

28. Nowicki M. Basic facts about biosimilars. Kidney

Blood Press Res. 2007;30:267–72.

29. Rathore N, Rajan RS. Current perspectives on stability

of protein drug products during formulation, fill and

finish operations. Biotechnol Prog. 2008;24:504–14.

30. Vulto AG, Crow SA. Risk management of biosimilars in

oncology: each medicine is a work in progress. Targ

Oncol. 2012;7(Suppl 1):S43–9.

31. European Medicines Agency. Guideline on similar

biological medicinal products. http://www.ema.

europa.eu/docs/en_GB/document_library/Scientific_

guideline/2013/05/WC500142978.pdf. Published May

22, 2013. Accessed July 19, 2013.

32. DrugBank. Acetylsalicylic acid. http://www.drugbank.

ca/drugs/DB00945. Updated February 8, 2013. Accessed

July 19, 2013.

33. DrugBank. Trastuzumab. http://www.drugbank.ca/

drugs/DB00072. Updated July 14, 2011. Accessed

July 19, 2013.

34. United States Food and Drug Administration. Guidance

for industry: scientific considerations in demonstrating

biosimilarity to a reference product. http://www.fda.

gov/downloads/Drugs/GuidanceComplianceRegulator

yInformation/Guidances/UCM291128.pdf. Accessed

July 19, 2013.

35. World Health Organization. Guidelines on evaluation

of similar biotherapeutic products (SBPs). http://

www.who.int/biologicals/areas/biological_therapeut

ics/BIOTHERAPEUTICS_FOR_WEB_22APRIL2010.pdf.

Accessed July 19, 2013.

36. Strober BE, Armour K, Romiti R, et al. Biopharmaceut-

icals and biosimilars in psoriasis: what the dermatolo-

gist needs to know. J Am Acad Dermatol. 2012;66:

317–22.

37. Sharma B. Immunogenicity of therapeutic proteins.

Part 3: Impact of manufacturing changes. Biotechnol

Adv. 2007;25:325–31.

38. European Medicines Agency. Guideline on immuno-

genicity assessment of monoclonal antibodies

intended for in vivo clinical use. http://www.ema.

europa.eu/docs/en_GB/document_library/Scientific_

guideline/2012/06/WC500128688.pdf. Published May

24, 2012. Accessed July 19, 2013.

39. Casadevall N, Edwards IR, Felix T, et al. Pharmacovi-

gilance and biosimilars: considerations, needs and

challenges. Expert Opin Biol Ther. 2013;13:

1039–47.

40. Department of Health and Human Services. Food and

Drug Administration. Guidance for industry: good

pharmacovigilance practices and pharmacoepidemio-

logic assessment. US Department of Health and

Human Services, US Food and Drug Administration,

Center for Drug Evaluation and Research, Center for

Biologics Evaluation and Research, Rockville, MD;

2005.

41. Schellekens H. Bioequivalence and the immunogenic-

ity of biopharmaceuticals. Nat Rev Drug Discov.

2002;1:457–62.

42. Casadevall N, Nataf J, Viron B, et al. Pure red-cell

aplasia and antierythropoietin antibodies in patients

treated with recombinant erythropoietin. N Engl J

Med. 2002;346:469–75.

Page 20: Developing Oncology Biosimilars: An Essential Approach for the Future

J. AbrahamS24

43. Locatelli F, Del Vecchio L, Pozzoni P. Pure red-cell

aplasia “epidemic”–mystery completely revealed? Perit

Dial Int. 2007;27(Suppl 2):S303–7.

44. Boven K, Stryker S, Knight J, et al. The increased

incidence of pure red cell aplasia with an Eprex

formulation in uncoated rubber stopper syringes.

Kidney Int. 2005;67:2346–53.

45. Macdougall IC, Roger SD, de Francisco A, et al. Antibody-

mediated pure red cell aplasia in chronic kidney disease

patients receiving erythropoiesis-stimulating agents: new

insights. Kidney Int. 2012;81:727–32.

46. Azevedo VF, Sandorff E, Siemak B, Halbert RJ. Poten-

tial regulatory and commercial environment for bio-

similars in Latin America. Value in Health Regional

Issues. 2012;1:228–34.

47. Mody R, Goradia V, Gupta D. How similar are bio-

similars in India? A blind comparative study. Pharma

Focus Asia. 2010;11. www.pharmafocusasia.com/

research_development/blind-comparative-study.html

Accessed July 20, 2013.

48. Li E, Hoffman JM. Implications of the FDA draft

guidance on biosimilars for clinicians: what we know

and don’t know. J Natl Compr Canc Netw. 2013;11:

368–72.

49. US Food and Drug Administration. Guidance for

industry: formal meetings between the FDA and bio-

similar biological product sponsors or applicants.

http://www.fda.gov/downloads/Drugs/GuidanceCom

plianceRegulatoryInformation/Guidances/UCM345649.

pdf. Accessed July 19, 2013.