biosimilars: assets and sustainability, between regulation and information

45
Biosimilars: assets and sustainability, between regulation and information Final Project Work, March 2016 Master in Pharmaceutical Marketing & Management Alma laboris S.r.l. Author: Jacopo Sacquegno

Upload: jacopo-sacquegno

Post on 23-Jan-2017

168 views

Category:

Healthcare


1 download

TRANSCRIPT

Biosimilars: assets and

sustainability, between

regulation and informationFinal Project Work, March 2016

Master in Pharmaceutical Marketing & Management

Alma laboris S.r.l.

Author: Jacopo Sacquegno

Index

1.1 What are biopharmaceuticals?

1.1.1 Some definitions and general features

1.1.2 Notable differences with traditional drugs

1.2 What are biosimilars?

1.2.1 Comparing generics and biosimilars

1.3 How are biosimilars authorized?

1.3.1 Bioequivalence and regulatory needs;

1.3.2 Interchangeability and substitutability for biosimilars;

1. A SCIENTIFIC APPROACH

1.1 What are biopharmaceuticals?

«Broad Biotech»

Pharmaceuticals that are biological in nature and manufactured by

biotechnology methods. This includes both products manufactured by

recombinant DNA technology (e.g., monoclonal antibodies and

recombinant proteins; involving genetic engineering) and products

naturally present in living organisms and extracted (e.g., proteins

and vaccines derived from non-engineered organisms as well as

blood/plasma-derived products)

1.1.1 some definitions and examples

«New Biotech»

Only those pharmaceuticals biological in

nature and manufactured using genetic

engineering (hence, it includes only

biological products such as monoclonal

antibodies and recombinant proteins).

• Been around for over 200 years;

• Revenues reaching about 160$bn/yr

“Any virus, therapeutic serum, toxin, antitoxin or analogous product

applicable to the prevention, treatment or cure of diseases or injuries of

man” (FDA)

“A medicinal product manufactured with one or more of the following

biotechnological techniques: recombinant DNA techniques, controlled

gene expression and antibody methods” (EMA)

“A product, the active substance of which is a biological substance. A

biological substance is a substance that is produced by or extracted from

a biological source […]” (AIFA)

Definitions by some regulatory authorities…

RECEPTOR CONSTRUCTS (fusion proteins), usually based on a naturally-

occurring receptor fused to the immunoglobulin frame through DNA

recombinant techniques. In this case, the receptor provides the construct with

detailed specificity, whereas the immunoglobulin-structure imparts stability

and other useful features in terms of pharmacology.

Biosynthetically produced substances that are (nearly) identical to the body’s own key signalling

proteins, including:

Hormones, chemicals transfer information and instructions between cells in animals and

plants (INSULIN, GLUCAGON, GROWTH HORMONE, GONADOTROPINS);

Cytokines, hormone-like molecules that can control reactions between cells (INTERFERON,

INTERLEUKINS, ERYTHROPOIETIN);

Enzymes, complex proteins that catalyse organic reactions (INSULIN, GLUCAGON, GROWTH

HORMONE, GONADOTROPINS);

Various blood factor, involved in coagulation (FACTOR VIII, FACTOR IX) and thrombolysis

(TISSUE PLASMINOGEN ACTIVATOR);

MONOCLONAL ANTIBODIES. These are antibodies produced by using

hybridoma technology (i.e. hybridizing antibody-producing spleen cells with

immortalized cells) or other methods that allow to obtain antibodies made

by identical immune cells all clones of an identical parent cells and bind the

same target, in contrast to polyclonal antibodies which are made from

several different immune cells. When used as biopharmaceuticals, they are

designated with the stem -mab.

Technology Mechanism of

actionIndications

1. Adalimumab

(Humira®)

Monoclonal antibody TNF antagonist Rheumatoid arthritis, ankylosing spondylitis, psoriatic arthritis,

psoriasis, ulcerative colitis, Crohn's disease

2. Etanercept

(Enbrel®)

Recombinant human TNF-

receptor fusion protein

TNF antagonist Rheumatoid arthritis, ankylosing spondylitis, psoriatic arthritis, psoriasis

3. Infliximab

(Remicade®)

Monoclonal antibody TNF antagonist Rheumatoid arthritis, ankylosing spondylitis, psoriatic arthritis,

psoriasis, ulcerative colitis, Crohn's disease

4. Insulin glargine

(Lantus®)

Recombinant insulin Insulin analogue Diabetes

5. Rituximab

(Rituxan®, MabThera®)

Monoclonal antibody CD20 binding and B-cell

elimination

Many lymphomas, leukemias, transplant rejection and autoimmune

disorders.

6. Bevacizumab

(Avastin®)

Monoclonal antibody VEGF-A antagonist Colorectal, lung, breast, renal and brain cancer; several eye disease.

7. Trastuzumab

(Herceptin®)

Monoclonal antibody HER2/neu receptor

antagonist

Breast cancer

8. Pegfilgrastim

(Neulasta®)

PEGylated form of the

Granulocyte colony stimulating

factor (GCSF)

Stimulates neutrophils blood

concentration

Fight infections in patients undergoing chemotherapy.

9. Ranibizumab

(Lucentis®)

Monoclonal antibody fragment VEGF-A antagonist Age-related web macular degeneration

10. Erythropoietin alfa

(Epogen®, Procrit®, Eprex®, ESPO®)

Recombinant human

erythropoetin

EPO analogue Anemia from chronic kidney disease, chemotherapy-induced anemia in

patients with cancer, Crohn's disease and ulcerative colitis and

myelodysplasia from the treatment of cancer

Top-10 biologic active substances for global sales in 2014

1.1.2 Notable differences with traditional drugs

Traditional drugs Biopharmaceuticals

Size Tens or hundreds of Da Hundreds of thousands of Da

Complexity Relatively simple Much more complex

Origins Standardized organic reactions Living Organisms

Stability Stable Not so stable

Production Synthesis is relatively cheap, can be standardised and

is easily replicable.

Synthesis is expensive and hardly reproducible

(“product is the process”); purification is not

standardisable.

Treatment Used to treat “traditional” diseases. Used to treat otherwise untreatable pathologies.

Adverse effects Predictable. Possible immunogenicity.

MCBs are under trade secret,

hence no MCB is equal to the

other and this influences the

final product. Cell banks are

constantly examined in order

to avoid contamination from

external agents.

After scaling up in increasingly bigger

volumes, culture is collected, active

principle is extracted and goes through

several steps of purification before being

ready.

The whole process is

much more complex and

expensive than what is

usually required for

traditional drugs. The

complexity of the process

makes the final product

totally dependent from

each step. The same

molecule obtained by

two different companies

can be different at the

end of the manufacturing

process (e.g. in its

tridimensional structure

and post-translational

modifications), leading to

significant alterations in

terms of effectiveness

and safety. Even

different batches of the

same product display a

certain degree of

variability.

After culture, the resulting product-

producing cells are generally aliquoted and

frozen, creating identical and ready to use

“MASTER CELL BANKS” (MCB) that can be

indefinitely preserved.

Each “master cell bank” is

then thawed and used up to

create aliquoted “WORKING

CELL BANKS” (WCB), which

functions as viable source of

cells for a production batch.

This system allows to ensure

an essentially indefinite

supply of the originally

developed production cells

for manufacturing purposes.

«The product is the process»

*Adverse effects

A peculiar feature of biopharmaceuticals is their possible IMMUNOGENICITY, i.e. the

capacity of inducing an immune reaction. Once in the body, these recombinant

proteins can be recognized as “non-self” by the organism, which in response produces

anti-drug-antibodies (ADAs) inactivating the therapeutic effects of the treatment and,

in rare cases, inducing adverse effects. One example of this is the aplastic anemy that

was associated with a particular form of recombinant erythropoietin (Eprex®). The

prediction of the immunogenic potential of novel protein therapeutics is thus a

challenge in biotherapy.

*Molecular size

Ibuprofen has only 13 carbon

atoms and a molecular weight of

about 206 Da;

Simvastatin has 25 carbon atoms

for about 418 Da;

Vancomycin has 66 carbon atoms

for about 1450 Da.

Human growth hormone is a protein

with 191 amino acids and a

molecular weight of 22 kDa;

Human erythropoietin is a

glycoprotein with 165 amino acids in

its mature form, weighing 34 kDa;

Immunoglobulins have molecular

weights in the range of 150kDa;

Biopharmaceuticals are designed to target precisely on a single structure

(protein, receptor, DNA sequence), increasing the effectiveness of the therapy

and reducing side effects at the same time.

Biopharmaceuticals have had a profound impact on many medical fields,

primarily rheumatology and oncology, but also cardiology, dermatology,

gastroenterology, neurology and others, adding major therapeutic options for

the treatment of many diseases, including some for which no effective

therapies were available, and others where previously existing therapies were

clearly inadequate.

Autoimmune diseases (e.g. rheumatoid arthritis, ankylosing spondylitis,

psoriatic arthritis).

Many form of tumors (e.g. breast, renal, liver, lungs, brain, colorectal),

including lymphomas and leukemias.

Chronic inflammatory diseases (e.g. Chron’s disease, ulcerative cholitis).

*Treatments

1.2 What are biosimilars?

The increasing development of sequencing technologies, molecular cloning

processes and in vitro biological production has allowed the production of

virtually any recombinant DNA-based biological substance for eventual

development of a drug. Monoclonal antibody technology combined with

recombinant DNA technology has paved the way for tailor-made and targeted

medicines. Gene- and cell-based therapies are emerging as new approaches.

Since many of the first generation biopharmaceuticals have been developed

in the 80’s, they are getting close to their patent’s expiration date; for some

of them, this date is soon to come or has passed by already. This allows any

biotech company other than the original creator of the expired biological

product to develop and market the same product on its own. After years of

debate, these products have been defined “BIOSIMILAR MEDICINES” by the

EMA. The term clearly distinguish them from what generics (“bioequivalent”)

are with respect to traditional drugs.

1.2.1 Comparing generics and biosimilars

Generics Biosimilars

Structure Identical to the reference product. Most probably not identical.

Production Manufacturing process is highly standardized and

generally the same as the originator.

Manufacturing process can be different from the one

of the originator (possible patent).

Analysis Analytical techniques can demonstrate identity with

the originator.

Analytical techniques cannot fully characterize

complex molecules.

Contaminants Detectable and quantifiable. Different depending on the substrate/organism used.

Formulation Rarely influences bio-equivalency. May interfere with therapeutic effects.

Safety & Efficacy variations Minimal. Cannot be inferred without supporting clinical trials.

Generic drugs are based on active

substances, which are bioequivalent to

another active substance already

authorized, with the same quali-

quantitative composition,

pharmaceutical form and therapeutic

indications. Mass spectrophotometry

and nuclear magnetic resonance can

safely identify the active substance

and detect contaminants;

manufacturing procedures, being highly

standardised, are generally the same

as those of the originator; different

excipients and final formulations have

minimal effects on the final safety and

efficacy of the drug.

A biosimilar and its reference product, on the

other hand, are obtained through inevitably

different manufacturing processes, which is due

to the inherent variability of the biological

expression system and the manufacturing

process itself. Differences in the former

determine differences in the contaminants, and

replication of the latter can be even harder

because the originator’s producer can hold

patent rights on the production process

itself. Moreover, analytic techniques cannot

fully characterize complex biological

molecules, which makes them insufficient to

demonstrate that the biosimilar product is

equal to its originator. An alternative final

formulation can also alter the biosimilar’s

properties.

“the absence of a significant difference in the rate and extent to which the active

ingredient or active moiety in pharmaceutical equivalents or pharmaceutical

alternatives becomes available at the site of drug action when administered at the

same molar dose under similar conditions in an appropriately designed study.” (FDA)

“[bioequivalency occurs if two medicinal products are] pharmaceutically equivalent

or pharmaceutical alternatives and if their bioavailabilities after administration in

the same molar dose are similar to such a degree that their effects, with respect to

both efficacy and safety, will be essentially the same” (EMA)

“[bioequivalency occurs if two medicinal products], at the same dosage, present

blood concentration profiles depending on time so similar that it is improbable they

will have relevant differences in their efficacy and safety effects”. (AIFA)

Definitions by some regulatory authorities…

Biosimilars cannot be completely bioequivalent due to the batch-to-batch variability

and their intrinsic biological nature. They are only “highly similar”, considering

minor differences in clinically inactive components and no clinically meaningful

differences in terms of safety, purity and potency.

In short, bioequivalent drugs must show the same absorption, the same upstream and

downstream blood levels, the same permanency in the blood circle and same excretion

time. These parameters define a drug’s BIOAVAILABILITY

The fraction of an administered

dose of unchanged drug that

reaches the systemic circulation

and it is described by the plasma

concentration time curve assessing

the rate and extent of absorption.

The area under the curve (AUC)

reflects the extent of exposure.

Cmax, the maximum plasma

concentration or peak exposure,

and the time to maximum plasma

concentration, tmax, are

pharmacokinetic parameters that

are influenced by absorption rate.

Most small molecule drugs are accepted as bioequivalent if

their AUC and Cmax are within the range of a 90%

Confidence Interval of 0,8-1,25.

Marketing authorization for

biopharmaceuticals is released by the EMA

through a Centralized Procedure.

The COMMITTEE FOR MEDICINAL PRODUCTS

FOR HUMAN USE (CHMP) writes the

EUROPEAN PUBLIC ASSESSMENT REPORT

(EPAR), published on the EMA website, which

includes the motivation for the favourable

response to the authorization request and a

section about presentation requirements

(labelling, packaging, etc.).

Attribution of a Community M.A. number.

Registration in the Community Register of

Medicinal Products (available on the

European Commission website).

1.3 How biosimilars are approved?

1.3.1 Regulatory needs and marketing authorization

EMA has been the first regulatory authority to

establish a specific set of rules for the approval

of biosimilar products. This legislative framework

later inspired several other countries (Australia,

Canada, Japan, Turkey, Singapore, South Africa,

Taiwan, the United States), as well as the World

Health Organization.

The authorization procedure follows EMA

guidelines:

General guidelines defining the concept of

biosimilars and establish general

requirements for authorization.

Guidelines for biosimilars’ quality;

Guidelines for preclinical and clinical studies;

Guidelines specific for each biosimilar line

(e.g., monoclonal antibodies,

erythropoietins, GH, ecc.).

“COMPARABILITY EXERCISE” is the

name for the overall process

aiming to ensure that the quality,

safety and efficacy of a

biopharmaceutical produced by a

changed manufacturing process

overlap the originator’s ones. The

exact requirements of

Comparability Exercises are set by

the EMA (in Europe) or the FDA (in

the U.S.). A Comparability Exercise

consist of a series of stepwise

procedures starting with tests for

quality (physical-chemical and

biological comparability) and

proceeding with non-clinical and

clinical trials (evaluation of safety

and efficacy).

CTD modules Generic drugs Biosimilar drugs

Module 1:

Administrative informationsComplete Complete

Module 2:

Summary of the next modulesComplete Complete

Module 3:

Product qualityComplete

Complete +

comparability exercise

Module 4:

Non-clinical reactionsOmissis / bibliographic

references

Preclinical tests results +

comparability exercise

Module 5:

Relations on clinical trialsBioequivalence study

Clinical trials results +

comparability exercise

Requirements for the Common Technical Document (CTD)

Step 1 - Quality comparability: this involves a thorough characterisation program comparing

the physicochemical and biological quality attributes, including the purity of the potential

biosimilar medicine as compared with the reference product.

Step 2 - Non-clinical comparability: As is the case with any biopharmaceutical, proposed

biosimilars must undergo pre-clinical studies before trials in humans are conducted. Data for

biosimilars are usually obtained via an abbreviated program of in vitro tests or studies in

animals, as required by EU guidelines. The non-clinical studies usually comprise repeat dose

toxicity, pharmacokinetic and pharmacodynamic studies in an appropriate animal model,

together with local tolerance testing in some instances.

Step 3 - Clinical comparability: A proposed biosimilar has to show that it is clinically

comparable to its reference biologic in terms of both efficacy and safety (phase I and III

clinical trials), including specific evaluation on immunogenicity. Specification of the clinical

information required takes into account existing comparability data, such as physicochemical

characteristics, receptor-binding assays and results from testing in animal models. The closer

the profiles of a biosimilar and reference products and the higher the similarity that has been

demonstrated through previous steps, the more an abbreviated clinical trial program can be

accepted by regulators.

Comparability exercise

1.3.2 Interchangeability & substitability for

biosimilars

“An interchangeable pharmaceutical product is one which is therapeutically

equivalent to a comparator product and can be interchanged with the comparator in

clinical practice.” (WHO 300 technical report series, no. 937, 2006)

“Interchangeability refers to the medical practice of replacing one drug with another

equivalent one in a given clinical context on the initiative or with the agreement of

the prescribing physician.” (European Generic Medicines Association, EGA)

“Interchangeability refers to the medical practice of replacing one drug with another

that has the same benefit-risk profile and is expected to have the same clinical effect

in a given clinical context on the initiative or with the agreement of the prescribing

physician.” (EFPIA/EBE-EuropaBIO)

Limits in the interchangeability of biosimilars

1. Manufacturing Process: reference biopharmaceuticals and their

biosimilar are similar but not identical. Therefore, it cannot be

assumed that they are automatically interchangeable. The switch

must be carefully monitored.

2. Lack of phase II clinical trials aiming to identify proper dosage:

clinical trials for the originator are taken into account.

3. Extension of indication: once the biosimilar’s effectiveness has been

demonstrated for one indication, it gets extended to all others. In

onchology, this could result inadequate, especially for monoclonal

antibodies. Each new therapeutic indication should be tested with a

specific registration process.

4. Immunogenicity risk: undetectable differences between biosimilars

and originators could lead to unexpected immunogenicity.

SUBSTITUTABILITY, on the other hand, refers to the practice of replacing a

drug with another drug, often at a lower cost for the Health Service or for the

patient, which has the same qualitative and quantitative composition of

active substances, the same pharmaceutical form and route of administration

and is bioequivalent to the reference medicine on the basis of appropriate

bioavailability studies. Automatic substitutability (of equivalents) by

pharmacists refers to the practice by which the pharmacist may, or must, in

accordance with national or local regulations, dispense a drug equivalent and

interchangeable instead of the prescribed medicine, without consulting the

prescriber. Finally, as regards substitutability, these differences should be

noted:

• primary substitutability refers to the medical practice of beginning a new

treatment with a biosimilar product (or an equivalent) rather than with

the originator reference product;

• secondary substitutability, on the other hand, refers to the medical

practice and/or of the pharmacist of modifying the treatment of a patient

already being treated with a biologic drug with its biosimilar.

2.1 How is the biopharmaceutical market?

2.1.1 The present-day global scenario

2.1.2 The lead actors on the biopharma market

2.2 How does biosimilars impact the market?

2.2.1 The past and the present of biosimilars

2.2.2 The future of biosimilars

2.3 How is the biosimilar situation in Italy?

2.3.1 The AIFA and the regional heterogeneity;

2.3.2 Interchangeability and substitutability for biosimilars;

2. A MARKETING APPROACH

2.1 How is the biopharmaceutical market?

2.1.1 The present-day global scenario

Sales from biotechnology

products have grown from a

share of 14% in 2006 to 23% in

2014, and they are expected to

reach 27% of the global market

by 2020.

Top-100 blockbuster drugs in 2006

comprehended only 21% biophar-

maceuticals; in 2014, this figure

has much increased reaching 44%.

The forecast for 2020 see the

percentage to increase to 46%,

lower than the 52% predicted in

last year’s report (which would

have meant a reversal in the

major market share of top selling

drugs between biological and

conventional medicinal

products).

Humira® (adalimumab)

was the single most

lucrative product in 2014

($12 billion).

Humira® generated $35

billion sales over the

period 2010-2013.

A total of 37 individual

biopharmaceuticals

recorded blockbuster

sales (>$ billion).

Taken together, the top

ten generated $69.8

billion sales in 2013

(50% of total

biopharmaceutical

revenues)

In terms of target indications, the majority of antibody and antibody-like products target inflammatory

and/or autoimmune conditions (cumulative 2013 sales of $41bn) and cancer (2013 cumulative sales of

$26bn). Among nonantibody–based products, insulins are the next most lucrative product class, collectively

generating sales of $21.5bn in 2013.

Number of biopharmaceuticals approved for market:

a. Approvals in the US and EU for each year from 2010 to 2013 period;

b. Approval numbers over the indicated period.

More than 250 biopharmaceutical products licensed in both markets over the considered period.

In 2014, 150 biologics were marketedworldwide and more than 370 were in the pipeline.

Product approvals as a percentage for each

class over the total, in the 2010-2014 period

and from 1982 to 2014 (cumulative).

There has been a decrement in approval for

some classes (such as interferon and

interleukins) in the last period, likely

reflective of market saturation.

Monoclonal antibodies, on the other hand,

have continuously grown in importance;

from about 10% of all approvals by the

end of the 1980s to 22% by 2014.

Promising research into synthetic biology and continuing computer-related progress in

biotech areas: Many bioinformatics studies are carried out for gene sequencing, drug

discovering, searching new drug receptors and producing tailored drugs which act specifically

on the target.

Unsatisfied therapeutic needs: biopharmaceuticals are the only known medications capable

of being used for the treatment of diseases and disorders toward which other therapies are

otherwise often ineffective.

Vast research investments by major pharmaceuticals firms: many major Pharmaceutical

companies are going to augment their biological portfolio and they have greatly increased

their research investments or benefitted from their partnering or acquisition of biotechnology

companies.

Major Pharmaceutical firms went or will go off patent: Many major conventional drugs,

including best-sellers like GSK’s Advair® (2010), Pfizer’s Lipitor® (2011), BMS’s Plavix® (2012),

Novartis’ Diovan® (2012) and AstraZeneca’s Nexium® (2014) have recently gone off patent, or

will do so in the near future, which means a significant boost to generic manufacturers that

quickly issues their own low-priced versions.

Reasons of this success

2.1.2 The lead actors on the biopharma market

Roche can count on three of the most important

anti-tumoral monoclonal antibody products in its

pipeline (Avastin®, Herceptin®, Rituxan®),

totalizing more than $20 billion revenues in 2014

taken all together.

By 2014, Amgen had 12 approved biological

medicines. Among these, the largest selling product

lines were Neulasta® and Enbrel® (co-marketed

with Pfizer in the U.S.)

Novo Nordisk, a diabetes specialist, manufactures

many recombinant insulin-based products (e.g.

Novolog®/Novorapid®, Levemir®,

Actrapid®/Novolin®).

Sanofi has recently seen a slowdown in Lantus®

sales, but it has enforced partnerships with the

American biotechnology firm Regeneron to develop

and launch monoclonal antibodies; four specialities

are already at a critical stage.

Originated in 2013 as a spin-off of Abbott

Laboratories, Abbvie owes most of its fortune to its

best-selling drug Humira® (almost 55% of total

revenue in 2014).

Mergers & Acquisitions

Pfizer acquired Wyeth

Biotech in 2009.

Producer of Enbrel®,

co-marketed with

Amgen.

Roche acquired

Genentech in 2009

Producer, among

others, of Avastin®,

Herceptin®, Rituxan®

and Lucentis®

...and also Sanofi + Genzyme ($20.1bn), the world third-largest biotechnology firm; Pfizer +

Hospira ($17bn), world's leading supplier of injectable drugs and infusion technologies; etc.

Merck & Co. acquired

Schering-Plough in

2009.

Producer of several

biologics, including

Remicade®

A wave of acquisition by

Big Pharma toward

successful biotech

companies, in order to

acquire biologic expertise

and market.

However, innovation in

biopharmaceuticals have

started to slow down

with respect to late

1990s (saturation for

therapeutic indication

and more restrictive

requests from regulatory

agencies may be the

cause)

Entering biosimilar

market may be an

alternative way to create

value with biologic for

Big Pharma.

2.2 How do biosimilars impact the market?

2.2.1 The past and the present of biosimilars

March 23th, 2012: the Obama administration

passed The Biologics Price Competition and

Innovation Act (BPCI Act), an amendment to the

Public Health Service Act to create an abbreviated

approval pathway for biological products.

July 24th, 2014: First biosimilar application to

FDA.

August 8th, 2014: First monoclonal antibody

application to FDA.

March 6th, 2015: Zarxio® by Sandoz is the first

biosimilar approved by FDA. Its reference product

is Amgen’s Filgrastim (originally licensed in 1991).

July 1st, 2005: First biosimilar application to

EMA.

April 12th, 2006: Omnitrope® (somatropin) by

Sandoz is the first biosimilar approved by EMA.

2010: By the end of the year, 14 biosimilars

were approved in EU, establishing a market that

reached about $172 million.

March 1st, 2012: First monoclonal antibody

application to EMA.

September 10th, 2013: EMA authorizes sale of

Remisma® (biosimilar of Remicade) by Celltrion

Healthcare in the EU.

Tim

elin

e

In Europe In the U.S.

Drivers

Growing pressure to reduce healthcare expenditure in developed countries;

Increase in the number of patients that can afford prescription drugs in developing markets;

Various blockbuster biologics going off patent;

High prevalence of chronic diseases in ageing population;

Regulatory initiatives (e.g. the EMA introduction of a licensing procedure; the BPCI act decided by the American government).

Restraints

High initial investment in research and development;

Medical efficacy and patient safety.

Stringent regulations in developed economies to restrict investments.

COSTThe average R&D investment for a

biopharmaceutical is $1.2bn. As a

consequence, for example,

Remicade costs almost $18.000 a

year; Herceptin costs up to $40.000

a year per patient. The BGMA

estimates that by 2020 biosimilars

could shave up to €33bn off drug

bills in eight EU countries.

TIMEIt takes an average of 10-15 years

to develop and market a new

biopharmaceutical. Biosimilars may

benefit from shortened licensing

process and previous research

studies. In this way, their marketing

time is reduced to 7-8 years.

After an initial burst of approvals

from 2006–2008, the EU approval

rate has slowed. This is hardly the

opening of the biosimilar’s

floodgate that someone predicted.

Market penetration was slow,

although now biosimilar market in

the EU stays at around $360mn,

with global total sales reaching

about $676mn (0.4% of the

biological market) (IMS Health).

On the positive side:

• by the end of 2012, 41% of EU

filgrastim market and 19% of

short-acting EPO market were

covered by biosimilars.

• Biosimilar prices in the EU have

an average 30% discount on the

product relative to their

originator.

• No unexpected safety issues

raised.

The European experience

An increasing number of

markets is developing their

own biosimilar pathways,

either based on EMA or

WHO.

Globally, by 2014 there

were 280 biosimilars in the

pipeline, and their clinical

trials is increasing by 20%

each year.

Two big markets like Russia

and China have still to

develop their own pathway.

As for biosimilar

manufacturing, India is the

third big player after EU

and the US.

The Asia-pacific region is

the most promising market,

with many biosimilars

already in use. By 2020, it

will become the highest

revenue generating area.

© 2016 - William Reed Business Media SAS - All Rights Reserved

Biopharma industry is in the middle of a

“patent cliff” in Europe and US, which

will put a market of more than $60bn

(considering only the top 10 selling

products) at risk.

The most important patent expirations

in the EU for the next future are

represented by Neulasta® ($4.4bn)

and, even more remarkable, Humira®

($12bn).

Biosimilars developers will be using

emerging markets with less intellectual

property protection to quickly enter

into established markets. Best example

for this is the Asia-Pacific Region:

• Collaboration between Mylan and

Biocon to commercialize biosimilar

of Herceptin® in India;

• Cadilla Healthcare launched in

India the first adalimumab

biosimilar under the name

Exemptia® in 2014.

2.2.2 The future of biosimilars© 2016 - William Reed Business Media SAS - All Rights Reserved

The challenge for biosimilar manufacturers is not only to implement a proper manufacturing

process, but also to prove comparability through clinical trials in a cost-effective manner. The

high upfront costs needed, both to manufacture and to execute clinical trials will, therefore, act

as a strong barrier to entry into the biosimilars market regardless of the ”patent cliff”.

The price drop for biosimilars will not be as pronounced as for generics, because companies

entering the market need to overcome these barriers yet still make a financial return (at date,

most biosimilars are sold at about 65%-85% of the originator’s price in the EU)

The key capabilities necessary to entry and prevail into the biosimilar market can be identified in:

• Research and Development: financial resources for basic research and clinical trials.

• Manufacturing: access to specific biomanufacturing facilities.

• Supporting activities: such as legal expertise and global distribution channels.

• Marketing: scientific information and a global network of sales representatives.

• Lobbying: with regulatory bodies, KOLs and governments.

2.3 How is the biosimilar situation in Italy?

2.3.1 Biosimilars impact on the Italian market

Therapeutic proteins

(growth factors,

interleukins, interferons,

insulins, ecc.) cover

almost all volumes of

biopharmaceuticals used

in Italy and more than

half of the expenses.

Monoclonal antibodies

represent only 5% of the

volumes, but this amount

translates into 42% of the

total expense for

biopharmaceuticals.Cesbio Bocconi – secondo rapporto di ricerca sui farmaci biotech (2014)

Cesbio Bocconi – secondo rapporto di ricerca sui farmaci biotech (2014)

By the end of 2013, 9 on 14

biosimilars approved by the

EMA were present on the

Italian market:

Omnitrope (somatropine) 29/10/07

Abseamed (EPOetin alfa) 03/11/08

Binocrit (EPOetin alfa) 29/10/07

Retacrit (EPOetin alfa) 03/11/08

Ratiograstim (filgrastim) 16/03/09

Tevagrastim (filgrastim) 01/11/09

Zarzio (filgrastim) 01/11/09

Nivestim (filgrastim) 08/06/10

Grastofil (filgrastim) 12/12/13

Italian market share of biosimilars has steadily grown.

Use of filgrastim biosimilars has become higher than

the reference product since 2012.

EPO biosimilars has sensibly grown as well, while

somatropine remained more or less stable.

Cesbio Bocconi – secondo rapporto di ricerca sui farmaci biotech (2014)

2.3.2 The AIFA and the regional heterogeneity

To date, the EMA has not expressed an official opinion yet about biosimilars’

substitutability, leaving decisional and legislative autonomy on the matter to

national authorities.

Recommendations suggest a precautionary approach:

In Italy, the AIFA makes it clear that biological medicines and biosimilars cannot be

considered purely and simply the same as equivalent products, and thus excludes

the mutual automatic substitutive therapy.

“The decision about the choice of prescribing a particular drug to be

used, rather than reference biosimilar, must be entrusted to qualified

health workers” (Ref. EMEA/74562/2006 Rev.1)

Tuscany (Regional Resolution n.592/2010)

In public purchase procedures for biosimilars,

composition, route of administration, therapeutic

indications and dosage must be indicated exclusively.

In no circumstance substitution with drugs equal in

composition, pharmaceutical form and dosage

produced by a different pharmaceutical company is

allowed.

Choice by the medical specialist to prescribe a drug

different from the one accepted by the public

purchase procedure must be motivated to the

people/person in charge of the institute of

reference.

Further costs deriving from prescription of drugs

different from those object of the public procedure

cannot be charged on the Regional Health System.

Molise (Regional Decree n.5/2010)

Biosimilar drugs must be used as first choice with

drug naïve patients, except for other therapeutic

indication and/or clinical judgement, which will be

object of a specific technical relation that must be

sent to the Therapeutic Regional Committee (CTR).

In patients already treated with biopharmaceuticals,

it must be guaranteed the alternative use of a

biosimilar, except for other clinical judgement or

welfare complexity conditions, object of a specific

technical relation that must be sent to the

Therapeutic Regional Committee (CTR).

Treatment with biopharmaceutical drug must be

guaranteed to the patient in case of therapeutic

inefficacy, non-sufficient therapeutic response or

manifested non-tolerance to the biosimilar.

Campania (Regional Decree n.34/2012) (D.n.44/2010 – n.15/2009)

All prescribing doctors, in the act of prescribing biopharmaceuticals, must show a preference to biosimilar drugs, with equal therapeutic indications and route of administration. Prescribers will motivate a different therapeutic choice with the specific patient card, which must be attached to the request and sent online, monthly, to the Nucleodi controllo dell’appropriatezza.

Patient card does not have to be filled for those biopharmaceuticals of the same fifth level-ATC category that got their price reduced with respect to the one established at the release date of Commissioner’s Decrees n.15 of 30/11/2009 and n.44 of 14/07/2010, allowing achievement of the expected 40% discount.

Concerning naïve patients, with equal therapeutic indication in the data sheet, the least expensive biosimilar must be used.

In case of documented therapeutic inefficacy and/or non-tolerance, use of another biopharmaceutical must be guaranteed.

Lacking a national policy about biosimilar’s substitutability, every Italian region has expressed or could express its own set of rules and measures, which can be discordant on the matter and put the equality of the citizen’s right to healthcare over all national territory at risk.

Tuscany, Molise and Campania are regions that have favoured biosimilar’s use, at least for naïve patients. For those being already treated with a biopharmaceutical, the principle of therapeutic continuity is generally held. Therapeutical switch toward a biosimilar must be carefully evaluated and decided only with the consent of prescribing doctor.

Nevertheless, for example, therapeutic continuity and personalization is more stressed in Tuscany’s resolution, while Campania’s regulations aim primarily to cut expenses. These are two different approaches that could probably lead to different guarantees for the citizens of the two regions.

Regions interested by resolutions on biosimilars and their relative number

over the course of the 2009-2012 period

Cesbio Bocconi – primo rapporto di ricerca sui farmaci biotech (2013)

(Re)defining biopharmaceutical (Rader, 2008; Nature)

Position paper of the Italian Society of Rheumatology on the Prescription of Biosimilars (2015)

Generici e biosimilari visti dal farmacologo (Rossi, 2013; 3^ conferenza sui farmaci a brevetto scaduto, Università di Napoli)

Farmaci biotecnologici e biosimilari - La rivoluzione dei biotech: dalle eritropoietine agli anticorpi monoclonali in oncologia (AIOM, 2013)

World Preview 2015, Outlook to 2020 (EvaluatePharma, 2015)

Biosimilars: Company Strategies to Capture Value from the Biologics Market (Fernandez & Hurtado, 2012; Pharmaceuticals)

Biopharmaceutical benchmarks (Walsh, 2014; Nature)

La diffusione dei farmaci biotecnologici, la loro costo-efficacia e i trend legati all'associazione farmaco-test per biomarker (Centro di Ricerche sulla Gestione

dell’Assistenza Sanitaria e Sociale, Università Bocconi, 2014)

BIBLIOGRAPHY (S0ME)