Sie sind auf Seite 1von 60

May 2017

Upholding the Clinical


Promise of Precision Medicine
Current Position and Outlook
Table of contents
Introduction 3

A systems approach to medicine 4

Systems therapeutics 4

Benefits of precision medicine 6

Characteristics of stratified and personalized medicines 7

Trends in the use of stratified medicines 12

Health system spending on stratified medicines 15

Novel therapies 20

Optimal use of systems therapeutics 22

Elements of an effective system for delivering full value from systems therapeutics 22

Use of big data and real world insights 26

Stakeholder roles 28

Challenges to improve the value of systems therapeutics for patients 31

Disparities in patient value 31

Variability of health outcomes 39

Recommendations for future applications of systems therapeutics 40

Multi-panel testing and increased use of next generation sequencing techniques 40

Sharing of healthcare data 41

Helping physicians make simpler decisions 41

Protecting patient privacy and data security 41

Evidence generation through increased collaboration 42

Notes on sources 43

References 44

Footnotes 50

Methodology 53

About the authors 58

About the Institute 59

2
Introduction
Recent advances in genomics, imaging and biomarkers are translating into the
development and use of systems therapeutics — medicines which target specific
pathways to modify the course of disease, and are intended for personalized or
stratified use in specific patient populations. Precision medicines are a subset within
systems therapeutics and have seen a period of rapid growth and acceptance
within healthcare systems, bringing the benefit of more effective treatments to
patients along with reduced adverse effects. This paradigm shift in medicine
will have resounding impacts on patients, while also transforming all aspects of
healthcare systems.

Challenges remain for health systems to maximize The study was produced independently by the IQVIA
positive health outcomes for patients from systems Institute for Human Data Science Institute as a public
therapeutics. Healthcare systems need to adapt to service, without industry or government funding. The
move efficiently from scientific breakthroughs to contributions to this report of Winfred Shaw, Terri
optimal patient care. As such, there is a growing Wallace, Amir Forouzan, Laura Mitrofan and others at
need to understand the requirements for systems IQVIA are gratefully acknowledged.
therapeutics to successfully reach patients and be
Find Out More
used to greatest effect.
If you wish to receive future reports from the IQVIA
This report introduces a framework for understanding Institute for Human Data Science or join our mailing list,
the unique challenges associated with systems visit iqviainstitute.org.
therapeutics including cost, clinical decision making,
MURRAY AITKEN
patient privacy, and the sometimes competing roles
Executive Director
of multiple stakeholders. In addition, case studies of
IQVIA Institute for Human Data Science
systems therapeutics provide insights into the lessons
learned on obtaining the optimal value of these
medicines. Finally, the report offers recommendations
for how systems therapeutics may achieve their full
potential for patient care in the future.

All analyses and conclusions are retained from this report previously published as the QuintilesIMS Institute. All references and citations to QuintilesIMS,
IMS Health or IMS data have also been retained to preserve the integrity of the report and analyses herein.

3
A systems approach SYSTEMS THERAPEUTICS
Systems therapeutics represent a paradigm shift in
to medicine drug development. Historically in medicine, drugs
primarily addressed disease symptoms, even if a
• Systems therapeutics are changing the medical molecular mechanism action was well understood.
landscape by directly targeting the biological systems However, the introduction of biologic treatments for
behind disease. multiple sclerosis and immune-mediated disorders in
the late 1990’s led for the first time to development of
• Precision medicine, a subset of stratified and
drugs that more specifically interfered with the disease
personalized therapies within systems therapeutics
process. The increasing application of systems biology
add significant value for patients and health systems by
in drug development led to further insights into the
offering improvements in response rate and tolerability.
mechanisms of diseases which in turn allowed for the
• Eighty stratified therapies require the use of predictive discovery of additional novel drug targets to interfere
biomarkers to identify patient response to therapies with and slow or halt the process of disease. In addition,
and they make up the bulk of systems therapeutics. new insights into the mechanisms of disease led to
further differentiation in diagnosis and treatments,
• Spending and volume of stratified medicines in
with significant advances in targeting patients with
oncology has increased dramatically over the past five
specific pharmacogenomic profiles for stratification
years with spending on stratified oncology medicines
of drug treatment. At the same time, novel insights
growing at a CAGR of 9.1% from 2011-2016.
into the mechanisms of inter-individual variation in
• The clinical trial landscape is increasingly including pharmacokinetics and pharmacodynamics have led to
the use of predictive biomarkers for patient selection personalized drug treatments.
with 350 studies selecting patients with predictive
Systems therapeutics are interventions which are:
biomarkers in 2016.
• Personalized with respect to the selection of
medicines and dosing

• Directly or indirectly impact disease progression

The introduction of biologic • Complex (incorporating patient status,


comorbidities, etc.)1
treatments for multiple sclerosis
and immune-mediated An understanding of systems biology has allowed
greater insight into the processes that govern disease
disorders led for the first time and has helped to usher in the era of precision
to development of drugs that medicine, where the right patient gets the right therapy

more specifically interfered at the right dose and benefits from the right response.
Systems therapeutics go hand in hand with precision
with the disease process. medicine in that they both include patient stratification
for drug response and personalization of medicines for
individual patients.

4
Predictive biomarkers are essential components in for testing of a specific gene, protein, or hormone
identifying those patients who can be stratified in prior to use in either the United States, Canada, Japan
regards to response and risk. Along with predictive or Europe. Products where testing of viral genotype
biomarkers, pharmacodynamic/pharmacokinetic was required prior to use were also included within
profiles of individuals are used to understand a the analysis. The 80 unique, stratified therapies in the
patient’s drug metabolism profile, anticipate drug– analysis span over eight broad therapy areas including
drug interactions, and will be necessary to track oncology, central nervous system (CNS), antivirals,
response to treatment. As a result, it will be necessary genetic disorders, inborn errors of metabolism,
to work with rational combinations of drugs and respiratory, immunology and rheumatology.
diagnostics to induce a change in disease processes,
Personalized medicine takes stratified medicine one
disease progression, and ultimately cure. In an ideal
step further as differential treatments tailored to
future, each patient may receive a unique treatment
individual patients based on their specific genome as
plan designed for them.
well as their status (e.g., pediatric, elderly, gender).
Systems therapeutics can be broken into two The goal of personalized medicine is to treat the
broad categories: stratified and personalized. Both underlying cause of the disease in the individual
stratification and personalization are components of patient (e.g., by altering the genetic code of the
precision medicine (see Exhibit 1, boxed area). Stratified individual), but, in addition, would include the use of
medicines provide differential treatments tailored pharmacogenetic screening to aid in predicting clinical
to specific groups of patients with individuals within outcomes for diseases with associated comorbidities,
the group receiving identical treatment. Stratified pharmacogenetic screening for drug–drug interactions
therapies for this report were selected for analysis in patients receiving multiple drugs, and identification
based on the following criteria: the medicines came of a patient’s unique pharmacokinetic profile to aid in
with a recommendation or requirement on their label drug selection and optimal dosing.

EXHIBIT 1: Systems Therapeutics

Pharmacologic Therapies • All symptomatic and disease modifying treatment

• Disease modifying therapies have an effect that modifies a disease t hrough an


Disease Modifying Therapies interaction with a specific target in the biological network u nderlying the disease
with the result being to directly or indirectly i mpact disease progression

• Differential treatments that are tailored to specific groups of patients based on


predictive biomarkers
• Individual patients in the patient group receive identical treatment
Stratified
Precision Medicines

• Not all stratified medicines are disease-modifying


-- Medicines stratified for dosing or adverse events recommendations d
 o not have
to be disease-modifying therapies

• Differential treatments that are tailored to individual patients based o n specific


genotypes as well as status (e.g., pediatric, elderly) or dose adjustments
Personalized • Can include cell-based or gene-based therapies
• Not all personalized medicines are stratified

Source: IQVIA Institute for Human Data Science, Apr 2017


5
BENEFITS OF PRECISION MEDICINE Overall, precision medicine can add significant
Precision medicines – medical treatment that is value for patients and health systems by offering
personalized to the characteristics of each patient by improvements in response rate and tolerability, dosing
stratifying individuals into subpopulations that differ guidelines, potential drug–drug interactions, and
in their susceptibility to a particular disease or their effective management of complex diseases. In addition
response to a specific treatment – have the potential to assessing treatment effect, pharmacogenomic
to provide significant value to patient outcomes. Prior testing can also identify those patients at greater risk
to the advent of precision medicines, all patients with for adverse events.2 Use and adoption of precision
a diagnosed disorder would receive similar treatment, medicine rely on the availability of predictive
with some individuals experiencing benefit and others biomarkers to identify patients who are susceptible
harm and added expense. The availability of precision to a particular drug effect, be it beneficial or harmful.
medicine now allows for personalized diagnosis Response to historical, first-line therapies across a
and treatment, with patients selected for treatment number of therapeutic areas are currently suboptimal,
based on individual traits and medicines developed and the ability to specifically target pathways in a
to specifically target the biological processes behind disease has the capability to offer improved efficacy to
diseases. A selection of the benefits of precision certain individuals who harbor specific genotypes.i,3,4
medicines is shown in Exhibit 2.

EXHIBIT 2: Examples of Success in Precision Medicine

• Chronic myeloid leukemia (CML) • A paradigm shift in the treatment of hepatitis C occurred through the use of
patients who have the typical direct acting antivirals (e.g., sofosbuvir, daclatasvir)
BCR-ABL1 fusion gene are treated • Success rates for the treatment of hepatitis C with DAAs reach more than 90%;
with a tyrosine kinase inhibitor (TKI) previously, success rates were approximately 50%
such as imatinib, dasatinib or nilotinib 
• Prior to the use of these Infectious
therapies, allogenic stem cell • Duchenne muscular dystrophy (DMD) is a rare, fatal
Diseases
transplantation was the only neuromuscular disorder
treatment for long-term control • 14% of patients with DMD have a confirmed mutation
• In one eight year follow up of the DMD gene that is amenable to exon 51 skipping
study, the estimated overall survival of • A recently approved therapy, eteplirsen, acts
all patients randomized to receive by restoring the translational reading frame
imatinib was 85% Rare
of DMD through specific skipping of exon 51
Diseases
• The overall survival of metastatic • Prior to the approval of eteplirsen, treatments of DMD
melanoma patients was less than 1 Oncology was symptomatic
year and long-term survivors were
• Cystic fibrosis is caused by mutations of the cystic fibrosis
rare prior to the introduction of
BRAF inhibitors transmembrane conductance regulator (CFTR) gene

• 57% of metastatic melanomas carry a mutation in the • CTFR modulators (e.g., ivacaftor and ivacaftor/lumacaftor) target
BRAF gene allowing targeted therapy with BRAF patients with specific mutations, such as G551D mutation or
inhibitor such as vemurafenib or dabrafenib F508del mutation

• Response rates for treatment with BRAF inhibitors in • Treatment was previously limited to symptomatic therapies
patients with specific BRAF mutations ranged from • Prior to the approval of CTFR modulators, treatments of cystic
48-59% in Phase II and Phase III clinical trials fibrosis was symptomatic

Sources: Blood 2009 114:1126; Immunotargets Ther. 2017 Feb 13;6:1-10; Ther Adv Med Oncol. 2016 Jan;8(1):48-56; Lancet. 2015 Mar 21; 385(9973): 1124–1135. For
full references see 5-8

6
The use of pharmacogenomic data to aid in treatment personalized genomic testing since the realization
decisions can also have a beneficial effect for other of the Human Genome Project have allowed for the
healthcare stakeholders. A study (n=110) conducted in development of methods to ensure drug response,
a hospital-based home health agency investigated the precise dosing, and minimize adverse drug reactions in
effect of using pharmacogenetic profiling along with individual patients.16
guidance from a clinical decision support tool (CDST) to
Predictive biomarkers allow the stratification of
aid in treatment decisions for patients receiving more
patient populations based on their predicted
than three therapies with the goal being to reduce the
response to a therapy. For example:
risk of dangerous drug–drug interactions. The results
of the study indicate that hospital readmissions and • Diagnostic tests identifying EGFR and ALK mutations
emergency department visits were decreased by 52% in non-small cell lung cancer, KRAS status in colorectal
and 42%, respectively, 60 days post-discharge in the cancers, and BRAF mutations in melanoma can help
pharmacogenetic tested group.9 The economic benefits stratify patient populations into those who will have a
from the use of precision medicine can therefore be more effective response to treatment.
consequential; the case study above lead to health care
• 57% of patients with metastatic melanomas carry a
savings of $4,382 per patient over 60 days.9
mutation in the BRAF gene allowing targeted therapy
The use of prognostic and predictive biomarkers to with BRAF inhibitor such as vemurafenib (Zelboraf)
facilitate disease prevention also has significant potential. or dabrafenib (Tafinlar); response rates for treatment
Cancer prevention programs can use pharmacogenetic with BRAF inhibitors in patients with specific BRAF
screening for early detection. Many neurological disorders mutations ranged from 48–59% in Phase II and Phase
are challenging to treat, and early identification remains III clinical trials.17,18
critical for optimal patient care and development of potential
Other predictive biomarkers can stratify patients
disease modifying treatments. In the future, it may be
according to risk. For example:
possible to identify risk of Alzheimer’s disease with predictive
biomarkers, and at present, biomarkers are increasingly • The availability of diagnostic tests to identify genetic
used in clinical trials for Alzheimer’s disease to support variants of the cytochrome P450 (CYP450) enzymes,
proof of efficacy. Research is also ongoing for prognostic
10 enzymes crucial for drug metabolism are significant
biomarkers to predict a risk for epilepsy and to improve tools to identify optimal drug dosing, inform risk of
multiple sclerosis diagnosis and disease progression. 11,12,13 adverse events, and predict drug–drug interactions.
Predictive biomarkers also provide an opportunity to identify
• Of the over 200 drugs on the FDA’s Pharmacogenomic
and stratify asthma patients, and in future, will help to predict
Biomarkers in Drug Labeling list, 35% of the
patient response to medication.14,15
biomarkers listed are for variations of a cytochrome
CHARACTERISTICS OF STRATIFIED AND P450 enzyme, and of those, 61% are for Cytochrome
PERSONALIZED MEDICINES P450 2D6 (CYP2D6).19 For the purpose of the analysis
Advances in the study of systems biology have for this report, we included those medicines that
transformed healthcare by moving the treatment required or recommended testing for the variant
paradigm away from symptomatic therapies to targeted on the label; however, labeling for CYP2D6 can be
disease pathways in specific individuals. In addition, informative and not require testing.ii,20
improvements in speed, access and affordability of

7
Characteristics of stratified therapies pharmacogenomic testing (see Exhibit 3). Thirty of
As of 2016, over 230 therapies across the United States, the stratified medicines in our analysis received a
Canada, Europe and Japan include pharmacogenetic pharmacogenomic biomarker that directed patient
information on their labels and the FDA’s list of stratification post-approval, and 40 have received
therapies with pharmacogenomic biomarkers in regulatory approval since 2011. The increasing
drug labeling totals over 200 medicines. 19,21
Stratified availability of therapies with pharmacogenetic
medicines included in the analysis in this report came information enables healthcare stakeholders to take
with a recommendation or requirement on their label actions to optimize the healthcare system to better
or based on regulatory professional society testing of a meet the needs of patients.
specific gene, protein, or hormone prior to use in either
Oncology contributed the greatest number of
the United States, Canada, Japan or Europe. Products
molecules and represented 58% of the total number of
where testing of viral genotype was required prior to
stratified therapies (see Exhibit 4).
use were also included within the analysis and these
included the direct acting hepatitis C therapies (e.g., • The increasing use of prognostic and predictive
sofosbuvir, daclatasvir). biomarkers to enable improvements in disease outcome,
effect of treatment, and reduction in risk of toxicity is a
A total of 80 therapies have been approved positive shift in the treatment of many cancers.
with labeling that requires or recommends

EXHIBIT 3: Number of Stratified Medicines

80 Total Number of Stratified Medicines Number of Stratified Medicines


by Biomarker Approval Date by Approval Type
70
9

60

50 7

40

5
30

20
2
10

0 0
0
1
2
3
4
5
6
7
8
9
0
1
2
3
4
5
6

6
7
0
5
7
9
200
200
200
200
200
200
200
200
200
200
201
201
201
201
201
201
201

201

201

201

201

201

201
197
199
199
199
199

Biomarker Post Approval Biomarker At Approval

Notes: Divalproex sodium and atazanavir were not included in the analysis; these drugs received regulatory approval in 1983 and 2003, respectively, but date of
biomarker approval could not be identified
Sources: https://www.pharmgkb.org/view/drug-labels.do; https://www.accessdata.fda.gov/scripts/cder/daf/;http://www.personalizedmedicinecoalition.org/
Resources/Personalized_Medicine_at_FDA; IQVIA Institute for Human Data Science, Apr 2017

8
EXHIBIT 4: Characteristics of Stratified Medicines

Therapy Area Year of Regulatory Approval Biomarker Function Black Box Status

6%
13% 18%

25%
45%
16%
58% 55%
69%
9%
83%

Oncology Genetic Disorders <1990 Efficacy BBW


CNS Immunology 1990-2005 Dosing, ADR or Both No BBW
Antiviral Rheumatology 2006-2016

100 Review Priority and Orphan Status

50 Priority
Standard

0 Orphan

Notes: ADR = Adverse drug reaction; BBW = black box warning; values may not add up to 100% due to rounding
Sources: https://www.pharmgkb.org/view/drug-labels.do; https://www.accessdata.fda.gov/scripts/cder/daf/; http://www.personalizedmedicinecoalition.org/
Resources/Personalized_Medicine_at_FDA; IQVIA Institute for Human Data Science, Apr 2017

• Targeted cancer therapies, including those therapies antiepileptic therapy drug class and biomarker testing
with predictive biomarkers as well as molecules that is recommended or required based on the potential
interrupt cell processes, such as angiogenesis (blood for adverse drug reactions.
vessel growth), represented 82% of all novel oncology
therapies launched from 2010 through 2014.22 Genetic disorders, which include inborn errors of
metabolism (e.g., hyperammonemia, Gaucher disease)
Sixteen percent of the stratified molecules consist and other genetic disorders (e.g., cystic fibrosis,
of antiviral therapies from the HIV and hepatitis C Duchenne muscular dystrophy), make up 13% of the
drug classes. number of stratified therapies.

• Globally, hepatitis C is associated with six main • Patient populations of these therapies tends
genotypes, each with different approved therapies to be minimal.
and response rates.
• For example, there are approximately 30,000
• Direct acting hepatitis C medicines are a success story cystic fibrosis patients in the United States and
for stratified medicines and provide high cure rates approximately 25,000 patients in Europe, while the
for hepatitis C. disorder is rare and under-reported in Asia.23,24

CNS therapies made up 9% of stratified medicines. • Even fewer patients have Gaucher Disease; in the United
States there are approximately 6,000 patients.25
• The bulk of these molecules belong to the

9
EXHIBIT 5: Components of Personalized Medicines

Therapies alter genetic information


in specific cells in individual patients
Individual Cell Line
Diagnostic testing can Therapies
ascertain the pharmacokinetic
profile of the patient to
aid in drug selection Dosing and
and dosing PK Effects Comorbidities Use of pharmacogenetic
screening can aid in
predicting clinical
outcomes in patients
with comorbid disease
Polypharmacy
Pharmacogenetic screening can help
identify drug-gene based interactions
in patients taking multiple therapies

Sources: PLoS One. 2017 Feb 2;12(2); IQVIA Institute for Human Data Science, Apr 2017

A singular characteristic of stratified medicines is that The regulatory characteristics of stratified medicines
there are a significant number of older, small molecules show that the majority of the therapies were approved
alongside more recently launched therapies and biologics. as new molecular or biological entities and 73%

• 80% of stratified medicines are small molecules. received priority regulatory review.

• 45% of stratified therapies are accompanied by a


• The greatest number of stratified therapies have been
black box warning.
approved or received biomarker designation post-
approval since 2006, yet 31% of stratified therapies • More non-orphan drug applications were associated
were approved before 2005 (see Exhibit 4). with a black box warning than orphan drug
applications (see Exhibit 4).
Diagnostic testing most often stratifies therapies based on
predictive effect with fewer therapies requiring diagnostic • Stratified therapies tend to treat serious, degenerative

testing to inform dosing and risk of adverse events. diseases (e.g., cancers) where providers and patients
are prepared to accept risks in order to slow or halt
• 83% of stratified medicines predict efficacy; within
disease progression.
this group there were four stratified medicines that
were associated with both efficacy as well as dosing • Patient outcomes will hinge, however, on safety as
and/or adverse reaction (see Exhibit 4). well as efficacy, and patient monitoring and support
during therapy is a key component of realizing the
• 18% of stratified therapies require testing
value of these therapies.
due to the prospect of adverse event or
dosing recommendations.

10
EXHIBIT 6: Stratified Therapies by Volume across the Developed Markets

Number of Number of Total Volume Growth 2006-2016 Growth 2011-2016


molecules molecules (Mn) 2016 %
2006 2016

All Stratified
Therapies 37 80 4,502 100% -7% -3%

CNS 6 7 2,698 60% -23% -12%

Oncology (total) 21 45 1,150 26% 40% 21%

Oncology
(excluding hormonal 14 38 216 5% 94% 32%
therapies)
Immunology/
Rheumatology 3 4 512 11% 37% 14%

Antivirals 4 14 139 3% -18% -10%

Genetic Disorders 3 10 4 0.1% 128% 87%

Sources: IQAVIA, MIDAS, Q4 2016; IQVIA Institute for Human Data Science, Apr 2017
Notes: Developed markets = United States, France, Germany, Italy, Spain, United Kingdom, Japan, Canada, South Korea, Australia; Oncology (excluding
hormonal therapies) does not include the following molecules: anastrozole, buserelin, exemestane, fulvestrant, letrozole, tamoxifen, toremifene; Count
includes Tositumomab iodine-131 and denileukin diftitox but no volume was available in 2016

Components of personalized medicines Gene therapies that treat non-oncology


Currently available personalized medicines are gene indications include:
therapies that can alter genetic information in specific • Alipogene tiparvovec (Glybera), an adeno-associated
cells in individual patients to treat or prevent disease. virus (AAV)-based gene therapy for the treatment of
Components of personalized medicine are shown in lipoprotein lipase deficiency (LPLD).
Exhibit 5.
• Strimvelis, a stem cell gene therapy for the treatment
Existing personalized gene therapies can be segmented of severe combined immunodeficiency due to
into oncology and non-oncology products. Examples adenosine deaminase deficiency (ADA-SCID).
within oncology include:
• Neovasculgen, a vascular endothelial growth factor
• Talimogene laherparepvec (Imlygic, formerly
(VEGF)-based gene therapy for the treatment of
OncoVex) a genetically engineered viral therapy for
atherosclerotic peripheral arterial disease (PAD),
the local treatment of recurrent melanoma.
including critical limb ischemia (CLI).
• Recombinant adenovirus–p53 (Gendicine), a viral
therapy for patients with a mutated p53 gene for the Though not a gene therapy, sipuleucel-T (Provenge) is a cell

treatment of head and neck squamous cell carcinoma. based immunotherapy for advanced prostate cancer that
modifies a patient’s personal immune cells with genetically
engineered fusion proteins which results in an increased
ability of the immune system to fight prostate cancer.

11
EXHIBIT 7: Oncology Stratified Therapies by Volume 2006–2016, Standard Units Mn

Oncology Units by Region Stratified Oncology Medicines Approved


1,200 10% 2011-2016

60
1,000
8%
50
800
6% 40
600
30
4%
400
20
2%
200 10

0 0% 0
6 7 8 9 0 1 2 3 4 5 6 1 2 3 4 5 6
200 200 200 200 201 201 201 201 201 201 201 201 201 201 201 201 201
ROW EU5 US % Unit Growth

Notes: ROW = Japan, Canada, South Korea, Australia


Sources: IQVIA, MIDAS, Q4 2016; IQVIA Institute for Human Data Science, Apr 2017

TRENDS IN THE USE OF STRATIFIED MEDICINES Use of molecules approved within the past five years
Total use of stratified medicines in the developed has grown through 2016 and has begun to level off
markets, measured in standard units, has remained (see Exhibit 6).
relatively flat since 2006. This is primarily due to the • Among the molecules with the highest volume since
higher representation of small molecule therapies their approval in 2011 are therapies from the oncology
within the group, many of which experienced generic and antiviral therapy areas, in particular, hepatitis C
competition within the study period, balanced against and chronic lymphocytic leukemia (CLL).
the launch of novel oncology and antiviral therapies
with low volumes (see Exhibit 6). Oncology
• 4.5 billion standard units were used by patients in the Use of stratified oncology therapies across the
10 major developed markets in 2016, out of 4 trillion developed markets has been growing since 2006 with a
units for global medicines.26 year over year growth rate of 4% in 2016 (see Exhibit 7).

• The top 10 oncology therapies make up 94% of the


• Within stratified medicines, older small molecules,
volume of stratified oncology therapies; all are small
particularly those in CNS and breast cancer, account
molecule therapies.
for over 75% of the volume.
• Tamoxifen, anastrozole and letrozole contribute the
• The perception that precision medicines are primarily
greatest share of volume and all are approved for
biologic, disease modifying therapies is balanced by
estrogen receptor (ER) positive breast cancer.iii,iv,27
the reality that there is also significant use of older,
Among the top ten molecules by volume, year over
small molecule medicines.
year growth rate of breast cancer was 4.2% in 2016.

12
• The oral chronic myeloid leukemia (CML) tyrosine • Overall volume of stratified antiviral therapies has
kinase inhibitors imatinib, ibrutinib and nilotinib also declined since 2006, due to the decline of the
contribute a significant amount to the volume of stratified HIV therapies as more patients move on to
stratified oncology therapies, although growth has newer medicines, such as dolutegravir, which is not
declined since 2010. Among the top ten molecules stratified (see Exhibit 8).
by volume, year over year growth rate of leukemia
• In 2016, direct acting hepatitis C therapies accounted
therapies was only 0.5% in 2016.v,28
for 32% of stratified antivirals.
• 48% of stratified oncology medicines received
• Hepatitis C therapies saw significant volume growth
regulatory approval since since 2011, and the CAGR
at time of launch, but uptake has since leveled off and
was 80% in the period from 2012-2016.
will no longer drive significant growth in this therapy
• Non-small cell lung cancer (NSCLC) was the only area in developed countries.
other therapy area represented in the top 10 oncology
molecules by volume due to use of erlotinib.
Antiviral therapies include those
Antivirals
HIV therapies stratified by risk
Antiviral therapies include those HIV therapies stratified
by risk of adverse events and the direct acting hepatitis of adverse events and the direct
C therapies that are stratified by viral genotype. acting hepatitis C therapies that
are stratified by viral genotype.

EXHIBIT 8: Antiviral Stratified Therapies by Volume 2006–2016, Standard Units Mn

Antiviral Units by Region Total Units, HIV and HCV, 2016


180 15.0%

160
10.0%
140
5.0%
120
32%
100 0.0%

80 -5.0%
60
-10.0%
40
68%
-15.0%
20

0 -20.0%
6 7 8 9 0 1 2 3 4 5 6
200 200 200 200 201 201 201 201 201 201 201
ROW EU5 US % HIV Unit Growth HIV HCV

Notes: ROW = Japan, Canada, South Korea, Australia


Sources: IQVIA, MIDAS Dec 2016; IQVIA Institute for Human Data Science, Apr 2017

13
EXHIBIT 9: CNS Stratified Therapies by Volume 2006–2016, Standard Units Mn

CNS Units by Region Total Units, CNS Drug Class 2016


4,000 10%

3,500 8%

6%
3,000
4%
2,500
2%
2,000
0%
1,500
-2%
1,000
-4%
500 -6%

-8% 98%
6 7 8 9 0 1 2 3 4 5 6
200 200 200 200 201 201 201 201 201 201 201

ROW EU5 US Spending Growth AED Other

Notes: ROW = Japan, Canada, South Korea, Australia


Sources: IQVIA, MIDAS Dec 2016; IQVIA Institute for Human Data Science, Apr 2017

CNS • The use of other CNS medications have declined in


Current therapies for CNS are stratified by risk of large part due to the launch and continued uptake
adverse events, in particular, for therapies where of more tolerable therapies, that, in turn, do not have
cytochrome P450 testing is recommended or required. labeling requirements for pharmacogenomic testing.
As a whole, these molecules are older, genericized and Though effective, the risk of adverse events and side
less tolerable therapies. effects lead these older, stratified medicines to be

• Total units have been declining since 2006; the year prescribed less often.

over year growth rate for 2016 was -2.3% (see Exhibit 9).

• Antiepileptic drugs (AEDs) make up 98% of stratified


CNS therapies.
Current therapies for CNS are
• Sodium valproate (divalproex semisodium; Depakote)
has the largest volume among the stratified CNS stratified by risk of adverse
therapies. The medicine has been launched globally events. As a whole, these
with multiple formulations and has approvals across
mood disorders, migraine and epilepsy. As such, the
molecules are older, genericized
volume of sodium valproate has remained relatively and less tolerable therapies.
stable since 2006.

14
EXHIBIT 10: Stratified Therapies by Value across the Developed Markets, 2006–2016

70

60

50
Spending US$Bn

40

30

20

10

0
2006 2007 2008 2009 2010 2011 2012 2013 2014 2015 2016

ROW EU5 United States

Notes: ROW = Japan, Canada, South Korea, Australia


Sources: IQVIA, MIDAS Dec 2016; IQVIA Institute for Human Data Science, Apr 2017

HEALTH SYSTEM SPENDING ON • Spending on stratified medicines in the EU5 reached


STRATIFIED MEDICINES $14.4Bn in 2016. EU5 countries experienced significant
Spending on stratified therapies in the 10 developed growth in the past five years, with a CAGR of 11.3%
markets totaled over $62.8Bn in 2016 (see Exhibit 10). over the period of 2011 through 2016. However, growth

• Spending across the developed markets increased at a declined from 2015–2016 by -8.1%. This decline was

CAGR of 20.5% from 2011–2016, driven primarily from due in part to the end of hepatitis C therapy uptake, as

growth outside of the United States and EU5; however, well as policymaker responses to unexpectedly high

growth slowed to 3% in 2016. This change in growth new drug spending in 2014 and 2015.

rate is largely driven by a reversal in hepatitis C-driven • Although overall spending was dwarfed by the United
therapy growth seen during the 2013-2015 period. States and the EU5, sharp growth was seen in the

• In the United States, spending on stratified therapies rest of the developed countries under study (Japan,

was $38.2Bn in 2016. Growth in the United States over Canada, South Korea and Australia) with a combined

the 2011–2016 period was also due to historically high CAGR of 19.3% over the 2011–2016 period.

price increases for branded and generic products


on an invoice price basis, in particular, the high
price points of oncology and hepatitis C medicines.
Spending on a net price basis – reflecting off-invoice
discounts, rebates and other manufacturer price
concessions – is estimated to be $30.5Bn.

15
Oncology and antiviral therapies made up the top five • Spending on biologics has increased from $10Bn in
stratified therapies by spending. 2011 to $14.9Bn in 2016. The greatest growth was

• The top five medicines by spending in the developed seen in small molecules, which accounted for $48Bn

markets included the oncology products imatinib, in spending in 2016.

rituximab, trastuzumab and the hepatitis C therapies • Overall, combined spending for stratified oncology
ledipasvir/sofosbuvir (Harvoni) and sofosbuvir products declined from a high of 77% of the total
(Solvadi) (see Exhibit 11). spending on stratified medicines in 2011 to 47% in

• Spending on breast and blood cancers remained 2016 due to the rapid uptake of hepatitis C medicines.

relatively flat from 2011–2016. Most notable are Antiviral agents accounted for 44% of spending on

the launch and uptake of ledipasvir/sofosbuvir and stratified medicines in 2016.

sofosbuvir, which offer essentially an effective cure for


hepatitis C. By 2016, these two medicines accounted
for 33% of total stratified medicines spending.

EXHIBIT 11: Trends in Stratified Medicine Spending across Developed Markets

Top 5 Product Spending by Disease Area


70
60 8.8
Spending US$Bn

9.2
50 4.5 4.4 Therapy Area, 2011 Therapy Area, 2016
40 9.4 20.6
4.4 23.7
30 10.7
20 8.3 8.5 8.9 3%
3.9 4.0 4.2 5%
10 6% 4%
0
2011 2012 2013 2014 2015 2016 11%
47%
Biologic and Small Molecule Spending
70
60 44%
Spending US$Bn

13.4 14.9
50 77%
40 12.6
30
11.3 47.5 48.0 100% = $24.7Bn 100% = $62.8Bn
20 10.0 10.5
32.5
10 14.8 15.0 16.6
0 Oncology Antiviral Immune/Rheumatology
2011 2012 2013 2014 2015 2016 CNS Genetic Disorders

Leukemias Breast Cancer HCV


All Others Biologics Small Molecules

Sources: IQVIA, MIDAS Dec 2016; IQVIA Institute for Human Data Science, Apr 2017

16
EXHIBIT 12: Oncology Stratified Therapies by Value 2006–2016

30 35%

25 30%

25%
20
Spending US$Bn

20%
15
15%
10
10%

5 5%

0 0%
2006 2007 2008 2009 2010 2011 2012 2013 2014 2015 2016

ROW EU5 US % Spending Growth

Notes: ROW = Japan, Canada, South Korea, Australia


Sources: IQVIA, MIDAS Dec 2016; IQVIA Institute for Human Data Science, Apr 2017

Oncology • Half of the top ten stratified oncology therapies are


Spending on stratified oncology products in the used to treat types of leukemia, four treat breast
developed markets grew at a CAGR of 9.1% from cancer and one treats lung cancer.
2011–2016 with growth rate of 12.7% from 2015 to 2016
• The greatest spending was on the biologic therapies
(see Exhibit 12).
rituximab and trastuzumab; however, small molecule
• Cancer remains among the leading causes of therapies imatinib and palbociclib (Ibrance) also
morbidity and mortality globally with over 14 million contributed significantly to spending.
new cases each year and 8 million resultant deaths
each year, projected to rise by about 70% through
2022 to 22 million.29

• Stratified medicines have had a significant impact on


the treatment of cancer as treatments options and
regimens are being tailored to individual patients by
targeting specific molecular pathways and harnessing
a patient’s immune system.

17
EXHIBIT 13: Antiviral Stratified Therapies by Value 2006–2016

30

25

20
Spending US$Bn

15

10

0
2006 2007 2008 2009 2010 2011 2012 2013 2014 2015 2016

ROW EU5 United States

Notes: ROW = Japan, Canada, South Korea, Australia


Sources: IQVIA, MIDAS Dec 2016; IQVIA Institute for Human Data Science, Apr 2017

Antivirals • In 2016, the spending on the top five antiviral


Spending on stratified HIV and hepatitis C medicines medicines was approximately 90% of the total
totaled $27.6Bn in 2016 with a CAGR of 58% from spending on stratified anti-invectives in the
2011–2016 (see Exhibit 13). developed world, due in large part to price. The cost

• Spending on stratified antiviral therapies spiked of stratified hepatitis C products is on par with some

in 2014 following the launch of stratified hepatitis oncology products.vi,30

C products in the developed markets. Previously,


growth of spending on stratified HIV medicines was
relatively flat.

18
EXHIBIT 14: CNS Stratified Therapies by Value 2006–2016

Spending by Region Spending by Therapeutic Class

3 30% 3

20% 2.5
2.5
10%
Spending US$Bn

Spending US$Bn
2 2
0%
1.5 1.5
-10%
1 1
-20%

0.5 -30%
0.5

0 -40% 0
06 07 08 09 10 11 12 13 14 15 16 06 07 08 09 10 11 12 13 14 15 16
20 20 20 20 20 20 20 20 20 20 20 20 20 20 20 20 20 20 20 20 20 20

ROW EU5 US % Spending Growth Antiepileptic Antipsychotic Other

Notes: ROW = Japan, Canada, South Korea, Australia


Sources: IQVIA, MIDAS Dec 2016; IQVIA Institute for Human Data Science, Apr 2017

CNS
Spending on stratified CNS therapies totaled just
Spending on stratified CNS
under $2Bn in 2016 with an 8.6% CAGR since 2011
(see Exhibit 14). therapies totaled just
• Overall, the launch of generic competitors to under $2Bn in 2016 with an
extended release valproate semisodium (divalproex) 8.6% CAGR since 2011.
led to a sharp decline in spending since 2008.

• The antiepileptic drug class accounts for the greatest


spending within CNS while the antipsychotic class has
remained relative static.

• The other CNS class, which includes


dextromethorphan/quinidine (Nuedexta
and tetrabenazine, has driven growth since
dextromethorphan/quinidine’s launch 2011 in
the United States for the treatment of
pseudobulbar affect.

19
EXHIBIT 15: Clinical Development with Biomarkers by Indication and Phase

Clinical Trials including Biomarkers Phase 0-Phase IV


Other Biomarker Trials
Predictive Biomarker Trials
1,200 1,091 40% % Trials with Predictive Biomarkers
1,027 957
1,000
30%
800
600 20%
400
10%
200
0 0%
2014 2015 2016

Clinical Trials Involving Predictive Biomarker Patient Selection, by Therapy Area, 2016 7%
Phase 1 Phase 2 Phase 3 Phase 4

150 134 19%


36%
100 79

50 38
20 15 14 11 10 9 7 6 6
0
gy ease logy CNS y r y y r r y
colo tor une Othe olog tolog scula sorde rolog
is
On us D ocrin
o
es pira y/Imm a lm rma iova ic Di ep h
R rg hth De Card enet
ctio End Alle Op
N 37%
Infe G

Notes: Other includes- Women’s health/sexual health, transplantation, gastrointestinal, orthopedics


Sources: Clarivate Analytics Cortellis, Feb 2017; IQVIA Institute for Human Data Science, Apr 2017

NOVEL THERAPIES Patient selection (or stratification) by a means of a


The late-stage pipeline for novel medicines is robust. predictive biomarker is the defining criterion for a
As of November 2016 there were 2,240 total active precision medicine or systems therapeutics. Predictive
therapies in late-stage development and an average biomarkers are related to therapeutic response in that
of 40-45 new active substances are forecasted to be they identify patients who are susceptible to a particular
launched per year through 2021. These medicines
26
drug effect, be it a response in terms of efficacy or in
will address significant unmet needs across therapy terms of adverse events. Other types of biomarkers
areas which include cancer, autoimmune diseases and include prognostic, diagnostic and pharmacodynamic.
diseases of the CNS. Although the new medicines
Among clinical trials with start dates in 2016, 350
will include conventional mechanisms of action, they
studies selected patients using predictive biomarkers
will also include novel, precise targeting of biological
out of a total of 957 studies that included biomarkers
processes within complex diseases.
(see Exhibit 15).

20
EXHIBIT 16: Emerging Therapies in Clinical Development in Trials Associated with Predictive Biomarkers

Number of Unique Molecules Emerging Therapies by Therapy Area, 2016

164
147 10%
3%
116 4%
4%

6%
55%
7%

11%

2014 2015 2016


Oncology Infectious Diseases Endocrinology CNS
Immunology Respiratory Dermatology Other

Note: Other includes- Women’s health/sexual Health, transplantation, gastrointestinal, orthopedics, ophthalmology, cardiovascular,
nephrology, genetic disorder Sources: Clarivate Analytics Cortellis, Feb 2017; IQVIA Institute for Human Data Science, Apr 2017

• In 2016, 4,726 clinical trials were started. Of these, • The greatest number of trials that include predictive
49% included some form of clinical biomarker. Of biomarkers is within oncology, followed by infectious
the total trials that included biomarkers, an analysis disease and endocrinology.
was conducted on those trials with biomarkers that
measured a therapeutic effect or toxic effect in Overall, there were 164 unique molecules being
patients, not healthy volunteers. In total, 957 trials investigated among the clinical trials that used
were analyzed to determine which studies selected predictive biomarkers to stratify patient population in
patients using predictive biomarkers. 2016 (see Exhibit 16).

• The number of emerging agents has been increasing


• The percentage of biomarker trials that selected
since 2014.
patients using a predictive biomarker has increased
from 18% in 2014 to 37% in 2016. • The greatest number of emerging agents is in
oncology (55%) followed by infectious disease (11%).
• In predictive biomarker trials with a start date in
2016, the greatest percentage was in early stage • Endocrinology and CNS made up 6 and 7% of unique
development, with 36% and 37% of trials belonging to molecules, respectively.
Phase I or Phase II development, respectively.

21
Optimal use of systems ELEMENTS OF AN EFFECTIVE SYSTEM FOR
DELIVERING FULL VALUE FROM SYSTEMS
therapeutics THERAPEUTICS
The increasing number of stratified and personalized
• Systems therapeutics have unique traits that the current medicines realized in the past few years is expected to
healthcare framework does not adequately address, continue based on the number of quality molecules in
such as access to and coverage of diagnostics. late-stage clinical development (see Exhibit 16). This has
important implications for patients, providers, payers
• The use of systems therapeutics will generate large
and pharmaceutical and diagnostic manufacturers alike.
quantities of data. Data standardizing, sharing and
The current healthcare framework will need to adapt to
analysis among stakeholders while protecting patient
support the increasing number of systems therapeutics
privacy and confidentiality would support greater
to help select the best treatment option for a particular
patient value.
patient. The current healthcare system and how it
• There are a number of key stakeholders in regards supports systems therapeutics is described in Exhibit 17.
to the use of systems therapeutics. Key roles for
stakeholders include coordinating a patient’s
care and pharmacovigilance.

EXHIBIT 17: How Healthcare Systems Support Systems Therapeutics

Patients eligible for treatment enter


the healthcare system

A subset of patients are identified via


predictive biomarkers

Multiple stakeholders coordinate to + +


enable patient access to the medicine

Stakeholders establish and implement


appropriate use for individual patients + +

RWE of patient outcomes is fed back


into the healthcare system
Source: IQVIA Institute for Human Data Science, Apr 2017

22
EXHIBIT 18: Systems Therapeutics Present Added Challenges to Health Systems

Predictive Timely Access Intricate and Evolving Big Data


Biomarkers to Therapies Treatment Protocols
• Patient identification • Timely access to • Protocols need to keep • RWE is more complex
via a predictive systems therapies up with scientific and includes large data
biomarker is required after diagnostic testing breakthroughs sets (e.g., genomics,
is necessary proteomics)
• Smaller patient pools • Intricate treatment
• For many cases, there decisions
• Diagnostic
is a greater emphasis
reimbursement can • Individualized
on cost, reimbursement
present challenges protocols are needed
and delivery
for each patient

Source: IQVIA, Apr 2017

Payers, providers and manufacturers alike have an • Use of a companion diagnostic is stipulated in the
interest in optimizing the healthcare framework to label of the associated therapeutic product and is
realize full value for patients from systems therapeutics. intended to identify patients who are most likely to
Unlike “one size fits all” therapies, systems therapeutics receive benefit or harm from that product.32,33
have unique traits that the current healthcare framework
• Although diagnostic tests can be developed in
does not adequately support (see Exhibit 18).
parallel with a systems therapeutics medicine they can
Identification of patients also be developed post-approval or re-purposed for
The use of predictive biomarkers is the first critical additional indications.
step for the identification of patients for a systems
therapeutic medicine and a biomarker is considered Coverage of diagnostic tests by payers is a bottle

predictive should the effect of treatment show a difference neck in terms of adequate patient value from systems

between those patients screened positive for a biomarker therapeutics since payer coverage of diagnostic tests

compared with those who show a negative screen.31 varies. Without full access to appropriate screening, the
full value of systems therapeutics cannot be realized, as
• A predictive biomarker may be available as a specific
inappropriate patients may receive these therapies or
diagnostic test (e.g., a companion diagnostic) which
eligible patients could be missed. vi,vii,34,35
provides diagnostic information for use with a
specific, corresponding therapeutic product.

• Types of predictive biomarker assays,


including companion diagnostics, consist of
immunohistochemistry assays, next generation
sequencing (NGS) techniques, and molecular
diagnostics, such as PCR or Sanger sequencing.

23
Accuracy of diagnostic testing is important for optimal Payers are increasingly requiring demonstration of
use of systems therapeutics because incorrect cost-effectiveness or additional clinical benefit when
interpretation of test results can lead to the wrong approving novel therapies. This underscores the
treatment for patients or could expose patients to importance of stakeholders coordinating at each step
greater risk.ix,36
for expectations for use by the patient, especially
for therapies that require diagnostic testing and
The smaller population size of patients eligible for
are associated with greater, up-front cost. Although
systems therapeutics can also lead to missed screening
payers cover a substantial number of rare diseases and
opportunities, should there be a lack of awareness or
oncology products, coverage in not universal across all
support of the testing among healthcare stakeholders.
health care systems.x
Additional hurdles for diagnostics include:

• Insufficient sample size and quality The cost of systems therapeutics, particularly for
oncology or rare diseases, can be a significant hurdle for
• Geographic availability for testing stakeholders. In particular, orphan drugs, for serious or
life-shortening or life-threatening diseases, can achieve
• Timely access to test results
significant prices per year. For example, Glybera is one of
• Appropriate use and interpretation of test results the most expensive medicines ever launched, with a list
by physicians price of €900,000 per treatment. There are a number of
factors that drive orphan drug prices which include:
Availability to patients
• Impact on patient lives
Optimal use of systems therapeutics will depend
in part on patient access to more complicated • The value proposition of the medicine
medicines. Importantly, timely access to systems
• Disease severity
therapeutics are critical, particularly for oncology
medicines, and there needs to be coordination among • Competition within the indication
stakeholders to move smoothly from companion
diagnostic testing and interpretation of results to • Prevalence of the indication (with higher drug costs

efficient access to these medicines. typically associated with lower prevalence)

Factors influencing patient access include: Although the high cost of systems therapeutics are
shared across stakeholders, inevitably there is greater
• Physical availability of the therapy in the
shifting of cost to patients. Branded therapies are
location required
getting more expensive on a list price basis, and
• Payer coverage although this can be mitigated by patient assistance
programs or insurance design, when patient costs are
• Cost
too high, patients may choose to not use the medicine.37
• Reimbursement
Distribution of systems therapeutics is also an integral

Payer coverage and reimbursement is an important part of systems therapeutics reaching patients.

component for access of the initial companion Wholesale distributors work closely with hospitals and

diagnostic screen and also remains an important pharmacies to ensure timely availability after diagnosis

step for patient access to the system therapeutic. and can help mitigate cost of therapies by allowing

24
BRAF/MEK with another targeted inhibitor (such as
CDK, PI3K, ERK or AKT inhibitors) and/or combining
There is a need to simplify BRAF inhibitors with an immunotherapy.39,40
treatment decisions for providers
In addition, coordinated care between all providers
in order to provide the greatest and the patient has the potential to improve adherence
value to patients. and compliance to treatment regimens. The full
value of human capital within the healthcare system
must be utilized in order to provide patients with the
hospitals and pharmacies to receive therapies at a coordinated care, education and support needed for
discount to manufacturer list prices and to allow these better patient outcomes.
facilities to keep a more modest on-hand inventory.
Distribution may also come with additional handling Monitoring use and measuring outcomes
requirements, more so than typical small molecules, or among patients
involve additional steps, such as compounding prior to Adequate systems for pharmacovigilance are required
patient use. to ensure patient value and to feed information back into
the healthcare system to support cost-effectiveness and
Appropriate use by physicians clinical benefit assessments of systems therapeutics.
The complexity of systems therapeutics requires
that protocols for use of systems therapeutics Patients should be monitored during and after
be consistently updated with the latest scientific treatment to ensure adequate safety and avoidance
information and tailored to individual patients to ease of adverse events of systems therapeutics and/or to
treatment decisions for physicians. There is a need to ascertain disease progression and clinical outcomes.xii,41
simplify treatment decisions for providers in order to • To reach the full potential for patient value for systems
provide the greatest value to patients. therapeutics, side effects of the medications must
also be monitored and managed to ensure patient
The use of stratified oncology medicines is particularly
satisfaction and adherence for optimal effect.
complex. Predictive biomarkers are essential for
treatment in many oncology areas, and a personalized • Even patients receiving oral oncology medicines,
protocol for each individual patient is becoming the though associated with high rates of adherence, may still
norm. This increased complexity can lead to difficult benefit from provider or other stakeholder follow up to
treatment decisions by physicians who must draw from monitor adverse events, side effects and to make sure
clinical experience, disease area guidelines, academic medicines are taken as prescribed; many of the current
research and payer prescription guidelines. tools for adverse event reporting and adherence are
patient-reported which can introduce errors.42
• One example of the complexity of treatment
• In addition, patients receiving medications outside
decisions can be taken from advanced melanoma
the hospital setting may “over-adhere” to their
cancer.xi, 38 A physician with a BRAF positive patient
regimen, continuing to take medications even in the
has a number of options to consider including:
presence of significant adverse events, thus lowering
immunotherapy, monotherapy BRAF inhibitor,
the value of the medicine to the patient.42
combination of BRAF/MEK inhibitor, combination of

25
Analyzing patient outcomes helps to provide essential
information on the use of systems therapeutics that
Real-world data provides
can be used to influence future treatments. However,
the volume of data from patients receiving systems researchers with a robust
therapeutics is often larger, and more complex, than pool of information and can
traditional medicines.
help to reveal when optimal
• The use of systems therapeutics will generate large
quantities of electronic medical and health records
and suboptimal patient care
including health information related to disease has occurred.
diagnosis, diagnostic testing and results, treatments,
outcomes, provider visits, and, potentially personal
genomic, or even proteomic profiles. are working or not working within the current
healthcare system, and will enable steps to be taken for
• These large data sets will need to be effectively
stakeholders to best meet the needs of patients.
protected and shared between stakeholders in order
to provide the feedback necessary to support the use • For example, next generation sequencing technologies

of these therapies in future patients. allow for timely and low cost sequencing of a
patient’s genome although this is accompanied by
USE OF BIG DATA AND REAL WORLD INSIGHTS a large volume of data (~100 gigabytes); analysis
In healthcare, transactional data is now more typically of this large dataset in conjunction with EHRs can
being collected digitally. More data is being captured pin-point diseases or predict drug interactions for
through Electronic Health Records (EHRs) than in the past, individualized diagnosis and therapy.47
as physicians move away from paper-based systems.
“Real-world data” provides insights into the efficacy
• A study in the United States in 2014 showed that EHRs
and safety of medical treatments in everyday practice,
were being used by 76% of non-federal hospitals and,
and across much larger populations, it can be used
in 2013, over 78% of office based physician practices
to improve the health system’s ability to save lives by
recorded health information in real time on patient
getting the right care to the right patient at the right time
health conditions, vital signs, medicine use and
and identify and address major healthcare gaps (see
laboratory testing results.43,44
Exhibit 19).48 Real-world data provides researchers with
• A study in Japan in 2011 showed that 62.5% of major a robust pool of information and can help to reveal when
hospitals had adopted EHRs and 62% of practitioners optimal and suboptimal patient care has occurred.
in Canada had adopted EHRs by 2013.45,46
While clinical studies are generally not large enough to

Systems therapeutics are unique in healthcare given the provide information to tailor treatment to specific patients

scale of information accompanying their use. Big data or narrow subpopulations, as is the case for patients

analytics within large, complex health care databases receiving systems therapeutics, the analysis of real-

can provide crucial knowledge and insights. Analysis world sources allows for clinical decision support (CDS)

of the big data associated with systems therapeutics by providing an analysis of smaller populations across

will improve the understanding of how these therapies various settings.49 Analysis of real-world data can provide
information within subpopulations to aid in choosing

26
the right treatments based on diagnosis, comorbidities, • Identifying and assessing sources and causes
current medications, genomic profile, diagnostic test of suboptimal use of medicines – by healthcare
results and any additional completed procedures. professionals and patients.

Big Data’s Role in Healthcare • Predicting disease, drug interactions or adverse


Big data can shape healthcare by: events based on genomic and/or proteomic profile

• Predicting demand and budgetary impact.


There are a number of challenges to applying big data
• Assessing value in terms of patient outcomes, total analytics to real world data to generate the greatest
system costs, satisfaction. value for healthcare (see Exhibit 20). Those particularly
relevant to systems therapeutics include:
• Supporting pharmacists in demonstrating their role
• Accessing, analyzing, standardizing, sharing and
in supporting patient care, including appropriate
storing genomic data in conjunction with EHRs.
use of medicines.
• Protecting patient privacy and confidentiality; this can
• Identifying responders/non-responders in real world
be accomplished via de-identifying patient data.
settings to tailor clinical treatment approaches.

EXHIBIT 19: Twelve Main Sources of Real World Patient Data

Clinical Outcome Assessments


Lab/Biomarkers Data
Wearables
Mortality Data
Consumer Data
Social Media Data
Registries

Pharmacy Data

Electronic Medical
and Health Records
Hospital Data

Prospective and Enriched Studies Claims Data

Source: Quintiles and IMS Serving RWI Together: IQVIA Institute for Human Data Science 2016

27
EXHIBIT 20: Approaches to Maximize the Value of Big Data in Healthcare

Increasing the availability and accessibility of high-quality non-identified patient-level data sources
Standardization of data and interoperability to allow meaningful analyses
Dara sharing and infrastructure projects to create larger or richer pools of data for research, i.e., view the health system in its entirety,
or all aspects of a given disease
Continued processing of patient health data into non- identifiable forms for research by health information owners
Ensuring availability of unique and new types of health information to contribute to healthcare understanding

Increasing the use of non-identified patient-level data sources to conduct research


Broaden use of non-identified patient data for research where possible, including at facilities that use patient identifiable information
for other purposes

Universally applying best-practice privacy and security standards


Acceptance of best-practice de-identification frameworks across the industry
Broad adoption of these frameworks by all stakeholders
Adherence to these best-practice frameworks to ensure patient privacy
Greater transparency and accountability by data processors about the frameworks they use and how they handle non-identified data

Strengthening the impact of evidence through increased collaboration


Connect healthcare stakeholders through data to speed consensus and remove barriers to action
Multi-stakeholder alignment (commercial/academic/government/provider) as partners contributing to medical, scientific and
statistical healthcare research

Creative and tangible ways to ensure the output from this data has greater effects downstream on patient treatment

Source: Aitken M, Nass D. Closing the Healthcare Gap: the critical role of non-identified information, IQVIA Institute for Human Data Science, 2015 Oct

STAKEHOLDER ROLES challenges with reimbursement and coordination with


Stakeholders within the healthcare system are those multiple clinicians (e.g., a cancer patient may be under
persons or groups that have an interest in clinical the care of an oncologist, general practitioner, pain
decision making with different view-points on how specialist). Patients may also need support from other
best to generate value within the healthcare system stakeholders regarding tolerability concerns, side
(see Exhibit 21). Each stakeholder has a unique role effects, and the potential for drug–drug interactions.
and set of needs for managing and dispensing
Payer
systems therapeutics.
Payers, which include both commercial and public
Patient insurers, play a substantial role in how the patient and
Patients, family, caregivers, advocacy groups and other other stakeholders (e.g., clinicians) make decisions
support networks seek straightforward, affordable about diagnostic testing and treatment choices. As
access to medicines and clinical care. Patients receiving the bulk of systems therapeutics are costly oncology
systems therapeutics may require additional support medications, payers increasingly require evidence
to ensure proper use of the medicine, given the of cost effectiveness and clinical benefits to ensure
complexity of treatment decisions as well as navigating placement on formulary and have an interest in ensuring

28
EXHIBIT 21: Key Healthcare Stakeholders Providing Clinical Care Decisions for Systems Therapeutics

Clinicians Therapeutic
Pharmacist & Diagnostic
Manufacturer

Regulator Patient

Hospital Wholesaler

Payer

Source: IQVIA, Apr 2017

that treatment guidelines are followed. Payers’ role in Research into comparative efficacy or patient-centered
systems therapeutics also extends to the diagnostic outcomes can help to guide the decisions of public
testing associated with these medicines. Payers also health programs, payer coverage, or public programs
seek to obtain the best prices for medicines and that aim to present the most accurate treatment
diagnostics in their negotiations with manufacturers. information for different diseases.50 Regulators also
provide a framework for pharmacovigilance that is
Clinician
essential in monitoring current patients receiving
Clinicians, which in the context of systems therapeutics
systems therapeutics and helping to determine
includes not only doctors, nurses but other health
cost-effectiveness and clinical efficacy of current
professionals, such as pathologists or radiologists
medications and diagnostics.
who interpret diagnostic test results, are at the center
of clinical decision making on behalf of the patient. Therapeutic and diagnostic manufacturer
Clinicians are involved in developing treatment protocols Both therapeutic and diagnostic manufacturers
based on clinical experience and academic research contribute to clinical decisions by being the entities
and guiding treatment decisions to comply with payer most familiar with their medicine or diagnostic. Both
prescription guidelines. Clinicians seek to strike a types of manufacturers sustain education and support
balance between satisfying and supporting patients with for healthcare professionals and patients. Manufacturers
the need to optimize the time spent per patient. also provide critical information on usage issues and
health outcomes. Manufacturers also set list prices;
Regulator
medicines which cure a disease or dramatically prolong
Regulatory approval bodies and policy makers seek
life are not necessarily expensive, but increasingly their
to ensure that treatment decisions are based on the
cost is coming under scrutiny by other stakeholders.
best available clinical evidence for efficacy and safety.

29
Novel, branded specialty products, (which include need store large quantities of specialty medicines for
oncologic, rare disease and hepatitis medicines) are on immediate use.
average 15–20 times more expensive than new, non-
Hospital
specialty brands.26 Manufactures are responding to
Hospitals, which include, community health
these high list prices by providing off-invoice discounts
centers, community hospitals and their professional
and rebates at approximately 30% in the United States
associations, provide a structure around which health
and roughly 17% in Europe and lower in the rest of the
care decisions are made. Hospitals manage the impact
world.26 Manufacturers must coordinate closely with
of systems therapeutics on the budget of the institution
other healthcare stakeholders, such as payers and
and also manage the workloads and cost of hospital
pharmacy benefit managers, in regards to pricing.
pharmacies and nursing support. Systems therapeutics
Wholesaler are often involved in complex treatment regimens and
The wholesaler and wholesale distributors play a large may require access to specialists, diagnostic testing
role in the safe and timely physical flow of systems and interpretation, and non-oral delivery methods
therapeutics. Wholesalers leverage the capabilities (e.g., infusions), all of which are supported within a
and infrastructure of a complex supply chain to provide hospital system. In addition, coordination and sharing
access to these complex medicines. Wholesalers will of EHRs and other data can be organized within hospital
purchase medicines directly from manufacturers and systems, and analysis of this data plays a significant
store them at distribution centers throughout a region; role around systems therapeutics, which can include a
clinicians than work with wholesalers to order and significant volume of data from diagnostic screening
receive the medicines they need for their patients. (i.e., genomic sequencing) as well as monitoring
Wholesale distribution provides value to manufacturers drug interactions of current treatments and planning
who do not have to devote financial resources to deliver coordinated care within the system.
medicines directly to providers. Wholesaler distribution
Pharmacist
also helps to provide value to pharmacies and hospital
Pharmacists, which include both professional
systems by ensuring quality and timely product
pharmacists who work in local and mail pharmacies,
availability, so that hospitals and pharmacies do not
compounding centers and hospital pharmacies, provide
value to clinical decisions for systems therapeutics
by understanding and managing the patient’s total

Manufacturers set list prices; drug profile. Pharmacists are also point of care
references for dosing and administration and can warn
medicines which cure a disease of potential side effects. Systems therapeutics may
or dramatically prolong life also be distributed by pharmacists through specialty
pharmacies, which can provide assistance to patients
are not necessarily expensive,
navigating reimbursement issues or have insight into
but increasingly their cost manufacturer rebate or patient assistance programs.
is coming under scrutiny by
other stakeholders.

30
Challenges to improve the still a number of challenges faced by health systems to
deliver the full value of systems therapeutics to patients.
value of systems therapeutics The framework within the healthcare system set up to
for patients deliver systems therapeutics to patients includes patient
identification, pharmacotherapy availability, appropriate
• Appropriate use and access to diagnostic testing use, and monitoring use and measuring outcomes. At
remain hurdles for systems therapeutics. For example, this time, the full value of use of systems therapeutics
results from a U.S. survey show that approximately has not been fully realized by patients. Exhibit 22
13% and 18% of NSCLC patients in did not receive illustrates the case studies presented in each section.
an EGFR test or ALK biomarker tests, respectively,
DISPARITIES IN PATIENT VALUE
despite guideline recommendations.
Patient identification with predictive biomarkers
• Systems therapeutics in the developing world also The role of stratifying patients with predictive
face a number of challenges, including the availability biomarkers is fundamental for the use of systems
of diagnostic testing and access, particularly those therapeutics but significant gaps remain around their
medicines associated with higher costs. use. In developing countries, access to predictive
diagnostic testing may be impossible, given limited
• Availability of systems therapeutics to patients is
resources and lack of infrastructure needed to support
varied across the world. 43% of countries included
testing and results interpretation.51 Even in developed
in the analysis did not have access to a systems
countries, the limited availability of genomic sequencing
therapeutics treating genetic disorders, which include
centers provides a ceiling to the amount of genomic
disease areas such as cystic fibrosis and Duchenne
testing that can be easily and quickly carried out.xiii
muscular dystrophy.
Demonstrating value to payers sufficiently to result in
• Cost can influence patient access to systems
their coverage of diagnostic testing and to physicians
therapeutics. Results from a U.S. survey covering
to result in routine screening of patients remains an
ten systems therapeutics indicate that 17–23% of
obstacle to patient access. Healthcare stakeholders
oncologists were not aware of their patients’ insurance
need to address the issue of patients not receiving the
coverage when prescribing the therapy, which can
recommended diagnostic tests.
lead to a greater burden on patients to navigate
insurance coverage and payments and potentially In oncology, although the uptake of diagnostic testing
restrict access. with predictive biomarkers has increased over the past
five years, there appear to be disease-specific barriers
• Hurdles around pharmacovigilance in the developing
to receiving diagnostic testing (see Exhibit 23).
world put the value of systems therapeutics at risk. Taken
as a whole, these drugs are associated with a higher risk • Survey results are from a pool of approximately 425
of black box warnings and adverse events, which could oncologists and show the highest rates of non-
lead to lower patient value if not properly monitored. testing in PD-L1 and KRAS testing for lung cancer,
at 55 and 52%, respectively. However, PD-L1 has not
There has been significant progress made around yet been incorporated into guidelines and KRAS
development and use of systems therapeutics, with testing recommendations are mixed without also
multiple stakeholders within healthcare systems actively testing of EGFR. 52
supporting and promoting their use. However, there are

31
EXHIBIT 22: Case Studies Demonstrating the Challenges to Provide Full Value of Systems Therapeutics

CASE STUDIES

Identification of Patients Patient identification with predictive biomarkers

Next generation sequencing availability

Availability to Patients Global availability of systems therapeutics

Variability in access and health outcomes

Payer coverage of branded systems therapeutics

Appropriate Use by Physicians Use of pharmacogenomic testing recommendations: oncology

Use of pharmacogenomic testing recommendations: CNS

Monitoring Use and Measuring Challenges in pharmacovigilance: HIV


Outcomes Among Patients Challenges in pharmacovigilance: hepatitis

EXHIBIT 23: Patients Who Did Not Receive Diagnostic Testing by Oncology Area and Type, 2016

PD-L1
KRAS
FISH
ROS-1
NRAS
ALK
EGFR
IHC
KRAS
17p
BRAF
ER
PR
0% 10% 20% 30% 40% 50% 60%

Lung Breast Colorectal Melanoma CLL

Notes: Number of patients covered in the survey: Lung = 19,793; Breast = 12,534; Melanoma = 2,885; Colorectal = 11,224; CLL = 8,056
Source: IQVIA BrandImpact, 2016; IQVIA Institute for Human Data Science, Apr 2017

32
• Guidelines recommend diagnostic testing of NSCLC Next Generation Sequencing Global Availability
patients with predictive biomarkers, in particular, Next generation sequencing (NGS) enables the
EGFR and ALK. 53
However, survey results show sequencing of a human genome, or specific areas of
that approximately 13% and 18% of patients in the interest, quickly and accurately, compared to historical
survey did not receive an EGFR biomarker test or sequencing that takes significantly longer and can only
ALK test, respectively. analyze smaller segments of DNA. One reason that
NGS has not been incorporated into routine clinical
• Of interest is that while the rate of testing patients for
care is restrictions around access. NGS often takes
KRAS, EGFR and ALK have increased since 2014, the
place at larger institutions with significant infrastructure
rate of positive tests has not increased substantially,
investments; smaller health centers have to send out
indicating that oncologists may be overtesting in
samples for sequencing and analysis. In the developing
certain patient groups.
world this becomes more problematic as access to
• Survey results show that for melanoma, where systems NGS facilities are limited. The table below illustrates a
therapeutics are the gold standard treatment for sample of countries by regions with access to genomic
patients with BRAF mutations, only 1.4% of patients sequencing centers mapped against GDP/capita. Many
do not receive BRAF testing. developing countries, such as India and Brazil, have
made significant advances in supporting genomic
• Overall, breast cancer had the lowest rate of non-
sequencing due to greater spending on research and
testing, with the exception of the FISH test for
development, but many other countries, especially
determining HER2 status of the tumor. Although
those in Africa, will need increased support in order to
FISH test is more accurate than IHC, it is less widely
provide services to their populations.
available for routine screening.

160,000

Nigeria
Population/center (000)

Egypt
Iran China
India
Kenya
8,000
Peru Saudi Arabia
Kazakhstan Colombia
Senegal Russia
Tunisia Brazil
Philippines Malaysia
South Africa
Argentina
South Korea US
Germany
UK
400
0 5,000 10,000 15,000 20,000 25,000 30,000 35,000 40,000 45,000 50,000

GDP/Capita USD

Note: The average number of sequencing centers is an approximate value and includes academic centers. The average number of sequencing centers is based
on an analysis from: Helmy M, Awad M, Mosa KA. Appl Transl Genom. 2016 Mar 10;9:15-9. The number of available high throughput sequencing centers was also
considered as a bottom limit in certain countries and used to estimate the availability in the United States. Country populations were taken from UN estimates.
GDP estimates were based on the World Band Open Data source. A full list of sources is listed in the references.51,54,55,56,57

33
EXHIBIT 24: Global Availability of Systems Therapeutics, 2015–2016

46 Oncology 14 Antiviral 13 Genetic Disorders 7 Central Nervous System 4 Immunology/Rheumatology

Canada
Russia
40 13 0 6 2
32 8 2 3 2

Kazakhstan
United States 19 5 0 1 1 South Korea Japan
44 13 8 7 4 33 6 4 5 3
China 36 9 4 3 3
Taiwan
India 19 2 1 2 2
Mexico 30 9 2 6 3
20 7 0 6 3
31 9 0 5 2 Malaysia Hong Kong
Thailand
27 5 0 3 3 32 9 1 4 3
24 6 0 4 2
Vietnam Philippines
Singapore
Brazil 16 6 0 2 1 24 0 0 4 1
30 8 0 2 3
30 7 0 5 2 Indonesia
15 0 0 4 1
South Africa
20 5 0 4 2 Australia
32 11 2 4 3

Note: Cholic acid and carglumic acid were not included in the U.S. country analysis
Source: IQVIA, MIDAS, 2016; IQVIA Institute for Human Data Science, Apr 2017

Availability of system therapeutics medicines that are found primarily in Europe (e.g.,
Global availability of systems therapeutics Glybera). 43% of countries included in the analysis
did not have access to a systems therapeutic within
The availability of systems therapeutics (stratified and
this category.
personalized medicines) in 2016 across 47 countries is
shown in Exhibit 24 and 25 revealing: • CNS therapies as a group were more readily available
than antiviral and oncology medicines, likely due to
• Developing countries had less access to systems
their generic availability.
therapeutics as a whole than developed countries,
particularly within more costly oncologic and antiviral • Western European countries are more likely to have
groups (which includes hepatitis C therapies). access to systems therapeutics than Eastern European
countries, particularly around the category of inborn
• Significant variability was seen among access to
errors of metabolism, respiratory and genetic disorders.
oncology medicines when considering developed
versus developing countries, likely due to cost of the • Fewer antiviral systems therapeutics are found in the
agents and infrastructure requirements. former Eastern Bloc countries: on average, patients
in the former Eastern Bloc countries have access to
• Inborn errors of metabolism, respiratory and genetic
seven antiviral systems therapeutics compared to an
disorders were included in a single category (genetic
average of 12 in western European countries.
disorders) and represent the most at risk group
of therapies outside of the developed world; this
category also includes the rare disease personalized

34
EXHIBIT 25: Availability of Systems Therapeutics within the EU and Eastern Europe, 2015–2016

46 Oncology 14 Antiviral 13 Genetic Disorders 7 Central Nervous System 4 Immunology/Rheumatology

Finland Netherlands Poland Serbia


35 12 1 3 3 37 10 5 5 3 29 7 1 5 2 22 3 0 2 1
Sweden Belgium Lithuania Croatia
39 12 5 4 3 35 10 4 5 3 25 9 1 3 2 31 4 1 2 3
Norway France Slovakia Bosnia
37 11 6 7 3 41 10 5 6 3 35 7 2 4 3 18 2 0 2 2
Denmark Portugal Hungary Czech Republic
35 12 4 4 3 38 10 4 5 3 32 9 3 4 3 31 6 1 4 3
Germany Spain Romania Slovenia
42 12 8 5 3 33 11 3 5 3 25 6 0 3 1 34 8 2 2 3
UK Switzerland Bulgaria Austria
40 12 6 7 3 34 11 0 3 3 31 6 0 3 1 39 12 5 4 3
Ireland Italy Turkey
32 12 0 6 3 35 10 6 5 3 34 6 0 4 3

Source: IQVIA Institute for Human Data Science, Apr 2017

Variability in access and health outcomes • When the authors considered a lower limit for the
Lack of access to cancer medications is a significant potential number of HER2 positive patients, the
barrier to improved health outcomes. The use and United States was the first country to reach optimal
procurement of trastuzumab provides a strong example level of procurement of trastuzumab for HER2 positive
of how limited access to systems therapeutics can be patients, while only two out of nine Eastern European
associated with negative health outcomes. Trastuzumab countries reached the optimal level of procurement of
is a highly effective and relatively safe therapy indicated trastuzumab by 2013.
for patients with HER2 positive breast cancer and is the
• The authors of the study point out that although rates
gold standard therapy in HER2 positive patients. Results
of cancer survival have improved in Europe since
from one study investigating the number of patients
1999, there is a survival gap between Eastern Europe
in need of treatment with trastuzumab and the level of
and Western Europe in a number of cancers, including
procurement of trastuzumab from 2001–2013 showed
breast cancer, due in part to lack of public funding for
a significant difference in the uptake of trastuzumab
more costly therapies, such as trastuzumab, screening
across the United States and Europe.
programs and delayed diagnosis.58,59
• Most Western European countries and the United
States procured sufficient amounts of trastuzumab
necessary to treat appropriate patients while in
Eastern European countries, there was significant
underuse until the end of the study period.58

35
EXHIBIT 26: Impact of Insurance on Oncologist Choice of Systems Therapeutic

Aware and patient’s insurance coverage


allowed me to prescribe my first choice

Not aware of patient’s insurance coverage

Aware but did not consider


patient’s insurance coverage

Aware and patient’s insurance coverage led me to


prescribe something other than my first choice

0% 20% 40% 60% 80%

CLL Brands NSCLC Brands

CLL brands included: NSCLC Brands included:


Imbruvica, Venclexta Tarceva, Xalkori, Gilotrif, Iressa, Zykadia,
Keytruda, Tagrisso, Alecensa

Source: IQVIA BrandImpact, 2016; IQVIA Institute for Human Data Science, Apr 2017

Payer coverage of branded systems therapeutics scientific findings. An example would be the use of
Exhibit 26 shows results from a survey conducted in Programmed Death receptor 1 (PD-L1) testing for the
approximately 400 oncologists in the United States treatment of NSCLC.
that demonstrates brand coverage for ten systems • Two systems therapeutics have regulatory approval
therapeutics in NSCLC and CLL. that target PD-L1, nivolumab (FDA approval 2015; EMA
• On average, oncologists indicate that in 69–75% approval 2016) and pembrolizumab (FDA approval
of their patients receiving a systems therapeutics 2015; EMA approval 2016).
medicine for NSCLC and CLL, they were aware of their
• Guidelines do not fully support PD-L1 testing, despite
patients’ insurance coverage and allowed to prescribe
the approval of PD-L1 targeting therapies for NSCLC, in
their first choice of therapy.
part due to the mixed clinical trial results in this patient
• However, 17–23% of oncologists were not aware of population and a lack of physician consensus.60,61
their patients’ insurance coverage, which can lead to
• Survey results from a pool of 406 oncologists covering
a greater burden on patients to navigate insurance
approximately 20,000 NSCLC patients indicate that
coverage and payments.
only 38% of their patients received a predictive

Pharmacogenomic testing recommendations are not biomarker test for PD-L1 in 2016 (see Exhibit 23).

always followed: oncology • In a separate survey conducted in Q4 2016, when


Appropriate use of systems therapeutics will depend asked why they did not order PD-L1 biomarker testing
on continued physician education on newly approved for NSCLC patients, 36% noted that it was because the
treatments and emerging mechanisms of action as testing was not recommended in the guidelines and
well as consistent updates to guidelines and hospital 24% of physicians noted the lack of consensus on PD-
protocols. Guidelines are at times behind the latest L1 testing.

36
EXHIBIT 27: PD-L1Testing for NSLC, 2016

2016 Testing for PD-L1 in NSCLC Top reasons for Not Testing PD-L1 for NSCLC, Percent of Physicians

Lack of reimbursement from payers

12% 12%
Insufficient tissue sample

PD-L1 testing is not available as part


21% of the standard panel of biomarker
tests for this tumor type

Lack of a requirement for PD-L1 testing in


the product prescribing information

55% Lack of consensus on whether or not


PD-1/PD-L1 inhibitors are more efficacious in
patients who over-express PD-L1

PD-L1 testing is not currently


recommended in major clinical guidelines
that I follow (e.g., ASCO, ASH, NCCN)
Positive Negative No Test Other 0% 20% 40%

Notes: Patients = 2,000; other contains results pending, inconclusive, and unknown
Sources: IQVIA BrandImpact, 2016; IQVIA Institute for Human Data Science Apr 2017

Pharmacogenomic testing recommendations are not serious adverse events at the expense of a more
always followed: CNS efficacious outcome.2
Inappropriate use of systems therapeutics can lead to
• Thus there is a gap between patients’ approval
adverse events.
and value for a more tolerable therapy and how
• Carbamazepine is a first-line anticonvulsant agent stakeholders are using these therapies in routine
with global regulatory approval for epilepsy clinical care.
and mood disorders but use of the therapy is
complicated by a risk of serious adverse, immune- Monitoring use and measuring outcomes
mediated, drug reactions. Patient-centered value of care is related to a number
of factors, including the side effect profile and risk of
• Although pharmacogenetic testing is available to
adverse events associated with the use of medicines.
stratify patients into high risk groups, uptake of
Healthcare stakeholders benefit by setting up systems
the test has not been widely adopted in certain
to ensure patientcentered, pharmacovigilance to
European populations. 2
improve adherence and to monitor for adverse events.xiv
• A survey conducted with epilepsy patients and
Pharmacovigilance is a necessary aspect of a healthcare
neurologists sought to determine the preferences of
system to understand and prevent future deaths
patients and neurologist to inform carbamazepine
from ADRs, and in terms of systems therapeutics,
pharmacogenetic testing and results showed that
pharmacovigilance is of even greater importance.xv,62
both patients and neurologists were in favor of the
test; in particular, patients valued a lower risk of

37
Monitoring for ADRs associated with these drugs closely In order to effectively treat HIV/AIDS in developing
is an integral part of providing full patient value for all countries, pharmacovigilance to monitor adverse drug
medicines, particularly systems therapeutics. reactions are critical. Patients who experience ADRs

• When including both personalized and stratified may under-dose medication, or fail to take medication,

systems therapeutics, 41% are associated with black leading to poorer health outcomes. However,

box warnings for serious adverse events and can pharmacovigilance in African countries remains poor

require specialized monitoring by trained clinicians. when compared to pharmacovigilance methods in


developed countries:
• 18% of systems therapeutics are stratified by risk of
• According to a study on pharmacovigilance in Uganda
adverse events.
and South Africa, both countries have the burdens of
limited funding, limited trained staff, and inadequate
Challenges in pharmacovigilance: HIV
training programs with the authors concluding that
A number of systems therapeutics are associated
there was not adequate capacity to monitor medicines
with adverse events in certain patient populations.
according to minimal WHO standards.66
This has real world consequences when applying
the use of these medicines in countries with • Only 7 and 8 staff members, respectively, are
suboptimal infrastructure for diagnostic testing and assigned in South Africa’s National Adverse Drug
disease monitoring. Event Monitoring Centre and Uganda’s National
Pharmacovigilance Center.
In developing countries, the rate of mortality for
communicable infectious diseases is high, and in
Challenges in pharmacovigilance: hepatitis
particular, for HIV/AIDS, 70% of worldwide cases
Globally, approximately two billion people are infected
occur in sub-Saharan Africa.63 ADRs in developing
with hepatitis B. The viral infection is associated with
countries can be exacerbated by higher prevalences
approximately 620,000 related deaths per year. In areas
of malnutrition, tuberculosis, and in the case of HIV
where the infection is highly endemic, the prevalence
patients, anemia. Approved treatments for HIV include
can range from 8–15%, in developing regions such as
the use of antiretroviral therapies, such as abacavir;
Sub-Saharan Africa, the Amazon Basin and Central
individuals with the HLA-B*5701 allele receiving
and Southeast Asia.67,68 The prevalence of hepatitis
abacavir are at a higher risk of hypersensitivity
C is lower; approximately 170–180 million patients
reactions.xvi,64
worldwide are infected with the bulk of infections
occurring in Asia and Africa.67 Co-infection of the
Data on adverse events from HIV therapies in Africa points
viruses is common, particularly in regions where there
to high numbers of ADRs compared to less than 10% of
is a high endemic prevalence of hepatitis B.xvii,69 Many
antiretroviral therapy patients enrolled in randomized trials
patients are unaware they are co-infected, as patients
having treatment-limiting adverse events.62,65
may have an inactive hepatitis infection where the
• 18% of patients from a study conducted in
individual may not experience symptoms and may not
Botswana experienced adverse events that required
know they are carrying the disease.
treatment modification.64
A paradigm shift in the treatment of hepatitis C
• 65% of patients from a study conducted in Kenya
occurred through the use of direct acting antivirals
experienced adverse events.64

38
(DAA; e.g., sofosbuvir, daclatasvir) which essentially Although the incidence of breast cancer is higher in
offer a cure for hepatitis C. The therapies are approved developed countries, the mortality rate is higher in
for use worldwide and are stratified according to developing countries; the WHO estimates that 324,000
viral genotype. However, the use of DAA hepatitis C deaths from breast cancer occur in developing countries
therapies is associated with a small but very real risk compared to 198,000 deaths in developed countries.72
of hepatitis B reactivation, enough that the FDA and
Hurdles to treatment include:
the EMA have added warnings that direct health care
professionals to screen and monitor for hepatitis B in all • Later presentation (tumors diagnosed at more
patients receiving direct acting treatments.xviii,70,71 advanced stages)

Assuming a 90% cure rate, 275,000 people living in low- • Lack of access to specialists
and middle-income countries have avoided cirrhosis
• Lack of access to radiography
and liver cancer based on access treatment in 2015.71
Although daclatasvir will soon be available as a generic • Higher prevalence of hormone receptor negative and
option in some countries, stakeholders will assess triple-negative cancers
value of these more expensive therapies based on
• High cost
safety as well as efficacy. In developing countries with
limited access to resources for pharmacovigilance and • Poor patient monitoring and follow upxix
diagnostic testing, stakeholders may restrict access to
DAA treatments for hepatitis C due to fear of ADRs from The overall result is poor outcomes for breast cancer
co-infection with hepatitis B, which would lead to fewer patients in developing countries, and in particular,
cures and a higher rate of complications and death. Africa. One study suggests that the five year survival
rate for breast cancer patients in Africa does not exceed
VARIABILITY OF HEALTH OUTCOMES 60% for any low and middle-income country on the
Management of cancers, viral infections and rare continent compared to a survival rate of 80–90% for
diseases can lead to significant disparity in the patients in the United States, Europe and Japan.73,74,75
developing world compared to developed countries.
Sub-optimal patient selection, availability, appropriate
use and monitoring patient outcomes can compound Recommendations for future
healthcare disparity around the systems therapeutics
applications of systems
that offer effective treatments in these therapy areas.
therapeutics
Breast cancer is an area with the availability of multiple
systems therapeutics, including the gold standard Healthcare systems need to adapt their existing

treatment, trastuzumab, which has caused a significant approaches to ensure that the maximum clinical and

shift in survival rates in developed countries. There is economic value of systems therapeutics is delivered.

a high necessity to screen patients who would best There has been growing recognition that systems

benefit from systems therapeutics, but a lack of access therapeutics, and indeed, precision medicines, offer

to diagnostics to aid in selecting the optimal treatment improvements to the status quo by targeting treatment

in developing countries. to the right patients and preventing waste on patients

39
who would otherwise not benefit or be harmed. access and acceptance of NGS among stakeholders,
However, to maximize the value of systems therapeutics, policymakers must invest and prioritize genome
further progress by stakeholders will need to come wide studies. Already a number of countries have
through the following approaches: invested in precision medicine initiatives that

• Multi-panel testing and increased access to next involve the widespread genetic screening of large

generation sequencing techniques. populations, such as the All of Us precision medicine


initiative in the United States, which seeks to recruit
• Sharing of healthcare data. one million participants to share their genomic and
other personal health data, and the United Kingdom’s
• Helping physicians make simpler decisions around
100,000 Genomes Project. Evidence of the utility
complex diseases.
and cost savings of NGS by allowing the fast and
• Protecting patient privacy and data security. accurate selection of an appropriate systems medicine
must also be demonstrated to payers and other
• Evidence generation through increased collaboration.
reimbursement authorities in order to increase the use
coverage and payments and potentially restrict access.
of NGS in routine clinical care.

MULTI-PANEL TESTING AND INCREASED USE OF SHARING OF HEALTHCARE DATA


NEXT GENERATION SEQUENCING TECHNIQUES The full value of systems therapeutics will require
The movement from single panel, diagnostic tests that a patient’s genomic information is analyzed
to multi-panel, diagnostic tests is an ongoing trend. along with other real-world health care data. This
There is the potential that multi-panel testing, at requires significant IT oversight, particularly over the
least in oncology, could exceed the use of single standardization of data so that it can be linked across
panel, diagnostic tests within the next five years.76 databases and analyzed. Currently, policy makers and
In addition, although the use of NGS techniques health information management (HIM) companies have
are on the rise, routine clinical practice remains an stepped up to help to regulate and provide solutions for
important milestone. NGS enables the sequencing of healthcare stakeholders and the use of HIM companies
a human genome, or specific areas of interest, quickly will only expand over the next five years.
and accurately, compared to historical sequencing
that takes significantly longer and can only analyze The complexity and size of genomic and healthcare
smaller segments of DNA. At this time, limitations data associated with patients receiving systems
around access to NGS centers, challenges around therapeutics presents an even greater challenge
reimbursement in certain countries, privacy, consent around the infrastructure needed to store and analyze
and handling the amount of data associated with these the associated data as well and how to share the
techniques remain a bottleneck for increased use and data with multiple stakeholders. Within the next ten
value of systems therapeutics. years, policy makers and the healthcare industry will
need to make efforts to ensure that standards around
The continued trend of multi-panel diagnostic testing oversight of data storage and data sharing occurs and
can be encouraged in order to obtain greater cost- that technology to deliver information simply to key
savings for stakeholders and also sample-sparing stakeholders exists.
for patients. Within the next ten years, to improve

40
Healthcare systems need to adapt their existing approaches to ensure
that the maximum clinical and economic value of systems therapeutics
is delivered.

HELPING PHYSICIANS MAKE SIMPLER DECISIONS PROTECTING PATIENT PRIVACY AND DATA SECURITY
Systems therapeutics invariably are used within complex In order to reach individualized medical treatments
disease areas, such as oncology, or in areas with smaller, for each patient, there needs to be acceptance and
highly specialized patient populations, such as rare trust from the general public around the concept of
diseases. Multiple interacting factors such as age, sharing genomic information for use in research studies.
gender, comorbidities, polypharmacy, management of Policy makers can encourage and normalize the use of
side effects and evolving scientific literature must be genomic research through government initiatives, but
incorporated to provide holistic care for the patient. in order to gain support from the public, individuals
must not only consent to the use of their genomic
Improved access by physicians to educational material
data, but must be assured that their personal genomic
(e.g., free CME courses, online videos) that provides
information is private and secure.
basic scientific background, summaries of recent
research findings, and supported protocols from Of note, the public is increasingly demanding access
professional societies for specific diseases around the to their personal genomic data, as the success of direct
use of systems therapeutics, could increase awareness to consumer (DTC) companies, such as 23andMe,
and reduce physician frustration around the use of these shows. Policymakers are also responding; in 2017 the
medicines. The goal being that by 2021, physicians FDA approved the first DTC genetic test to estimate a
using systems medicines will be able to make faster, patient’s risk of disease. However, if increased used of
accurate and well-informed decisions around these genetic testing is to approach the majority of patients in
complex therapies. developed countries by 2021, patient consent, privacy
and data security will have to be more adequately
Improved communication between hospitals,
addressed. Although patient data can be de-identified
professional societies and academic researchers is also
to increase anonymity, multiple data bases are used
necessary to provide protocols to physicians for systems
when analyzing health care data, and this can allow
medicines that reflect the latest science, available
for third-parties to potentially re-identify patients,
treatments and management of potential side effects.
thus necessitating strong data security. Physicians and
Communication with diagnostic companies around
researchers must also obtain informed consent from
the use and interpretation of diagnostic tests will also
patients and explain how their data will and will not be
reduce the burden on physicians to understand and
used. In a recent case between Google’s DeepMind and
interpret complex test results.
the UK’s NHS, more than half a million patients in the

41
U.K. did not know their information was being shared
with Google to build an app.77 Lack of public support
for wide-scale genomic studies could significantly affect Healthcare stakeholders,
how future systems medicines are developed and used including policy makers, will
in routine clinical care.
need to coordinate at all stages
EVIDENCE GENERATION THROUGH INCREASED
of systems medicine, from
COLLABORATION
Fully incorporating new information into clinical practice development and approval,
from the use of systems medicine, such as improved through clinical protocols,
survival rates, long-term safety and adverse events, can
pharmacovigilance and post-
take decades. In addition, even when evidence comes
from randomized controlled trials or real-world data, marketing studies in order to
research findings may be ignored or simply fail to be ensure that criteria collected
recognized and have downstream impact. At this time,
there is limited evidence that precision medicine and as
and analyzed is sufficient to
such, systems therapeutics, have dramatically improved demonstrate value to all parties.
clinical outcomes.78 Economic assessments around the
cost savings of these medicines is also mixed, in part
due to the complexities around diagnostic testing.76 parties. A first step towards this type of engagement is
Another confounding factor in demonstrating the true occurring at present, as policy makers and commercial
value around systems medicines is that the gold standard companies (e.g., Verily/Google’s Project: Baseline) are
for evidence generation is the randomized-controlled investing in precision medicine and genome study
trial; for evidence generation, these trials would likely initiatives. However, other health care stakeholders,
be larger, costly, post-marketing studies that would be such as payers, therapeutic and diagnostic
challenging to enroll due to the inherent nature of the manufacturers, and physicians, will also need to be
smaller patient populations associated with systems engaged to ensure that evidence on how they define
medicines. Evidence generation for systems medicines value is also generated.
will likely require the use of novel trial designs, such as
the N-of-1 trials, that in aggregate can offer information
on how to better treat patient sub-populations.79

Healthcare stakeholders, including policy makers, will


need to coordinate at all stages of systems medicine,
from development and approval, through clinical
protocols, pharmacovigilance and post-marketing
studies in order to ensure that criteria collected and
analyzed is sufficient to demonstrate value to all

42
Notes on sources
This report is based on the IQVIA services detailed below. Analyses exclude OTC products and focus on
prescription-bound products. Spending is reported at wholesaler invoice prices and does not reflect off-invoice
discounts and rebates.

IQVIA MIDAS™ is a unique data platform for assessing worldwide healthcare markets. It integrates IQVIA National
audits into a globally consistent view of the pharmaceutical market, tracking virtually every product in hundreds
of therapeutic classes and providing estimated product volumes, trends and market share through retail and non-
retail channels. MIDAS data is updated monthly and retains 12 years of history.

IQVIA BrandImpact™ oncology data is based on a proprietary mobile research model and longitudinal
network of more than 400 i-enabled oncologists and is the only source of continuously-captured physician
treatment decisions for the biopharmaceutical industry. The real-time data generated by its information panel
of oncologists enables BrandImpact to provide unique insight into physician behavior and the influences on
that behavior. The combination of its network-generated syndicated data with its custom research and analytics
expertise enables BrandImpact to deliver more informed and actionable solutions to its customers’ critical
business issues.

43
References
1. 6th FIP Pharmaceutical Sciences World Congress (PSWC). Stockholm, Sweden. 2017 May 21-24.

2. Powell G, Holmes EA, Plumpton CO, Ring A, Baker GA, Jacoby A, et al. Pharmacogenetic testing prior
to carbamazepine treatment of epilepsy: patients’ and physicians’ preferences for testing and service
delivery. Br J Clin Pharmacol. 2015 Nov;80(5):1149-59.

3. Xalkori. Prescribing information. Available from: http://labeling. pfizer.com/ShowLabeling.aspx?id=676

4. Perjeta. Prescribing information. Available from: https://www.gene.com/download/pdf/perjeta_


prescribing.pdf

5. Deininger M, O’Brien SG, Guilhot F, Goldman JM, Hochhaus A, Hughes TP, et. al. International Randomized
Study of Interferon Vs STI571 (IRIS) 8-Year Follow up: Sustained Survival and Low Risk for Progression or
Events in Patients with Newly Diagnosed Chronic Myeloid Leukemia in Chronic Phase (CML-CP) Treated
with Imatinib. Blood 2009 114:11.

6. Force J, Salama AK. First-line treatment of metastatic melanoma: role of nivolumab. Immunotargets Ther.
2017 Feb 13;6:1-10

7. Eroglu Z, Ribas A. Combination therapy with BRAF and MEK inhibitors for melanoma: latest evidence and
place in therapy. Ther Adv Med Oncol. 2016 Jan;8(1):48-56.

8. Webster DP, Klenerman P, Dusheido GM. Lancet Seminar – Hepatitis C. Lancet. 2015 Mar 21; 385(9973):
1124–1135.

9. Elliott LS, Henderson JC, Neradilek MB, Moyer NA, Ashcraft KC, Thirumaran RK. Clinical impact of
pharmacogenetic profiling with a clinical decision support tool in polypharmacy home health patients: A
prospective pilot randomized controlled trial. PLoS One. 2017 Feb 2;12(2).

10. Hampel H, Frank R, Broich K, Teipel SJ, Katz RG, Hardy J, et. al. Biomarkers for Alzheimer’s disease:
academic, industry and regulatory perspectives. Nat Rev Drug Discov. 2010 Jul;9(7): 560-74.

11. Koepp MJ. The help of biomarkers in the prevention of epilepsy. Lancet Neurol. 2016 Jul;15(8):782-4.

12. Pitkänen A, Löscher W, Vezzani A, Becker AJ, Simonato M, Lukasiuk K6, et al. Advances in the development
of biomarkers for epilepsy. Lancet Neurol. 2016 Jul;15(8):843-56.

13. Housley WJ, Pitt D, Hafler DA. Biomarkers in multiple sclerosis. Clin Immunol. 2015 Nov;161(1):51-8.

14. Zissler UM, Esser-von Bieren J, Jakwerth CA, Chaker AM, Schmidt-Weber CB. Current and future
biomarkers in allergic asthma. Allergy. 2016 Apr;71(4):475-94.

15. Chiappori A, De Ferrari L, Folli C, Mauri P, Riccio AM, Canonica GW. Biomarkers and severe asthma: a
critical appraisal. Clin Mol Allergy. 2015 Oct 1;13:20.

44
16. Zhang G, Nebert DW. Personalized medicine: Genetic risk prediction of drug response. Pharmacol Ther.
2017 Feb 14.

17. Force J, Salama AK. First-line treatment of metastatic melanoma: role of nivolumab. Immunotargets Ther.
2017 Feb 13;6:1-10.

18. Eroglu Z, Ribas A. Combination therapy with BRAF and MEK inhibitors for melanoma: latest evidence and
place in therapy. Ther Adv Med Oncol. 2016 Jan;8(1):48-56.

19. FDA. Table of Pharmacogenomic Biomarkers in Drug Labeling. Updated 2016 Nov 7. Available from:
https://www.fda.gov/ Drugs/ScienceResearch/ResearchAreas/Pharmacogenetics/ ucm083378.htm

20. Bertilsson L, Dahl ML, Dalén P, Al-Shurbaji A. Molecular genetics of CYP2D6: clinical relevance with focus
on psychotropic drugs. Br J Clin Pharmacol. 2002 Feb;53(2):111-122.

21. PharmGKB. Drug labels. Updated 2016 Dec 8. Available from: https://www.pharmgkb.org/view/drug-labels.do

22. Aitken M, Kleinrock M. Global oncology trend report. IQVIA Institute for Human Data Science. 2016 Jun.

23. Cystic Fiboris News Today. Cystic Fibrosis Statistics. Available from: https://cysticfibrosisnewstoday.com/
cystic-fibrosis-statistics/

24. European Lung Foundation. Cystic Fibrosis. Available from: http:// www.europeanlung.org/en/lung-
disease-and-information/lungdiseases/ cystic-fibrosis

25. National Organization for Rare Disorders. Gaucher Disease. Available from: https://rarediseases.org/rare-
diseases/gaucherdisease/

26. Aitken M, Kleinrock M, Nass D. Outlook for global medicines through 2021. IQVIA Institute for Human Data
Science. 2016 Dec.

27. Centers for Disease Control and Prevention. Global Cancer Statistics. Available from: https://www.cdc.gov/
cancer/ international/statistics.htm

28. National Cancer Institute. Chronic Myelogenous Leukemia Treatment (PDQ®)–Health Professional Version.
Available from: https://www.cancer.gov/types/leukemia/hp/cml-treatmentpdq# cit/section_1.3

29. World Health Organization. Cancer. Fact sheet N°297. Updated 2017 Feb. Available from: http://www.who.
int/mediacentre/ factsheets/fs297/en/

30. Bedi, J, Evans J, Kumar S, Yadav A. Global Market Overview: Chronic Viral Hepatitis. IQVIA Disease
Insights. 2016 Sep.

31. Ballman KV. Biomarker: Predictive or Prognostic? J Clin Oncol. 2015 Nov 20;33(33):3968-71.

45
References
32. FDA. Companion Diagnostics. Updated 2016, Oct 5. Available from: https://www.fda.gov/MedicalDevices/
ProductsandMedicalProcedures/InVitroDiagnostics/ucm407297.htm

33. European Commission. Revisions of Medical Device Directives. Accessed 2017 Mar. Available from:
http://ec.europa.eu/growth/ sectors/medical-devices/regulatory-framework/revision_en

34 Cohen JP, Felix AE. Personalized Medicine’s Bottleneck: Diagnostic Test Evidence and Reimbursement. J
Pers Med. 2014 Apr 4;4(2):163-75.

35. Bücheler M, Brüggenjürgen B, Willich S. Personalised Medicine in Europe – Enhancing Patient


Access to Pharmaceutical Drug- Diagnostic Companion Products. Updated 2014 Nov. Available
from: http://www.epemed.org/online/www/content2/104/107/910/ pagecontent2/4339/791/ENG/
EpemedWhitePaperNOV14.pdf

36. Li M, Yu T1, Hu YF. The impact of companion diagnostic device measurement performance on clinical
validation of personalized medicine. Stat Med. 2015 Jun 30;34(14):2222-34.

37. Aitken M, Kleinrock M. Medicines Use and Spending in the U.S. IQVIA Institute for Human Data Science.
2016 Apr.

38. Eroglu Z, Ribas A. Combination therapy with BRAF and MEK inhibitors for melanoma: latest evidence and
place in therapy. Ther Adv Med Oncol. 2016 Jan;8(1):48-56.

39. Manzano JL, Layos L, Bugés C, de Los Llanos Gil M, Vila L, Martínez-Balibrea E, et al. Resistant mechanisms
to BRAF inhibitors in melanoma. Ann Transl Med. 2016 Jun;4(12):237.

40. Flaherty KT, Infante JR, Daud A, Gonzalez R, Kefford RF, Sosman J, et al. Combined BRAF and MEK
inhibition in melanoma with BRAF V600 mutations. N Engl J Med. 2012 Nov 1;367(18): 1694-703.

41. Essential Medicines and Health Products Information Portal: A World Health Organization resource. The
Importance of Pharmacovigilance - Safety Monitoring of Medicinal Products. Updated 2002. Available
from: http://apps.who.int/medicinedocs/ en/d/Js4893e/5.html

42. Patel K, Foster NR, Farrell A, Le-Lindqwister NA, Mathew J, Costello B, et al. Oral cancer chemotherapy
adherence and adherence assessment tools: a report from North Central Cancer Group Trial N0747 and a
systematic review of the literature. J Cancer Educ. 2013 Dec;28(4):770-6.

43. Charles D, Gabriel M, Searcy T. Adoption of Electronic Health Record Systems among U.S. Non-Federal
Acute Care Hospitals: 2008-2014 Quick Stat #47. Updated 2015 Apr. Available from: http://healthit.gov/
sites/default/files/data-brief/2014HospitalAdop tionDataBrief.pdf

44. Hsiao CJ, Hing E. NCHS Data Brief No 143, Use and Characteristics of Electronic Health Record Systems
Among Office-based Physician Practices: United States, 2001–2013. Updated 2014 Jan. Available from:
http://www.cdc.gov/nchs/data/ databriefs/db143.htm

46
45. Takabayashi K, Doi S, Suzuki T. Japanese EMRs and IT in Medicine: Expansion, Integration, and Reuse of
Data. Healthc Inform Res. 2011 Sep;17(3):178-83.

46. Chang F, Gupta N. Progress in electronic medical record adoption in Canada. Can Fam Physician. 2015
Dec;61(12): 1076-84.

47. He KY, Ge D, He MM. Big Data Analytics for Genomic Medicine. Int J Mol Sci. 2017 Feb 15;18(2).

48. Aitken M, and Gorokhovich L, Advancing the Responsible Use of Medicines: Applying Levers for Change.
IQVIA Institute for Human Data Science. 2012 Sep.

49. Aitken M, Nass D. Closing the Healthcare Gap: The Critical Role of Non-Identified Information. IQVIA
Institute for Human Data Science. 2016 Oct.

50. Agency for Healthcare Research and Quality: The Effective Health Care Program Stakeholder Guide.
Updated 2014 Feb. Available from: https://www.ahrq.gov/research/findings/evidencebased- reports/
stakeholderguide/chapter3.html#Stakeholders

51. Helmy M, Awad M, Mosa KA. Limited resources of genome sequencing in developing countries:
Challenges and solutions. Appl Transl Genom. 2016 Mar 10;9:15-9.

52. Lindeman NI, Cagle PT, Beasley MB, Chitale DA, Dacic S, Giaccone G, et al. Molecular testing guideline for
selection of lung cancer patients for EGFR and ALK tyrosine kinase inhibitors: guideline from the College
of American Pathologists, International Association for the Study of Lung Cancer, and Association for
Molecular Pathology. J Mol Diagn. 2013 Jul;15(4):415-53.

53. Novello S, Barlesi F, Califano R, Cufer T, Ekman S, Levra MG, et al. Metastatic non-small-cell lung cancer: ESMO
Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2016 Sep;27(suppl 5):v1-v27.

54. United Nations. The World Population Prospects: 2015 Revision. Available from: http://www.un.org/en/.
development/desa/ publications/world-population-prospects-2015-revision.html

55. Next Generation Genomics: World Map of High-throughput Sequencers. Accessed 2017 Mar. Available
from: http:// omicsmaps.com/

56. GOLD: Genomes OnLine Database. Accessed 2017 Mar. Available from: https://gold.jgi.doe.gov/

57. World Bank Group. World Bank Open Data. Accessed 2017 Mar. Available from: http://data.worldbank.org/

58. Ades F, Senterre C, Zardavas D, de Azambuja E, Popescu R, Piccart M. Are life-saving anticancer drugs
reaching all patients? Patterns and discrepancies of trastuzumab use in the European Union and the USA.
PLoS One. 2017 Mar 14;12(3).

47
References
59. De Angelis R, Sant M, Coleman MP, Francisci S, Baili P, Pierannunzio D. Cancer survival in Europe 1999-
2007 by country and age: results of EUROCARE--5-a population-based study. Lancet Oncol. 2014
Jan;15(1):23-34.

60. Novello S, Barlesi F, Califano R, Cufer T, Ekman S, Levra MG. Metastatic Non-Small-Cell Lung Cancer:
ESMO Clinical Practice Guidelines. Ann Oncol. 2016 Sep;27(suppl 5):v1-v27.

61. Grigg C, Rizvi NA. PD-L1 biomarker testing for non-small cell lung cancer: truth or fiction? J Immunother
Cancer. 2016 Aug 16;4:48.

62. European Commission. Proposal for a regulation amending, as regards pharmacovigilance of medicinal
products for human use. Regulation (EC) No 726/2004. Impact assessment. 2008. Available from: http://
ec.europa.eu/health/files/pharmacos/ pharmpack_12_2008/pharmacovigilance-ia-vol1_en.pdf.

63. World Health Organization. Global Health Observatory data: HIV/ AIDS. Accessed 2017 Mar. Available
from: http://www.who.int/ gho/hiv/en/

64. Nwokike J. Monitoring Adverse Drug Reactions in public health programs: the case of the Nigeria TB
program. Submitted to the U.S. Agency for International Development by the TBCAP Project. 2008.
Available from: http://apps.who.int/medicinedocs/ documents/s18400en/s18400en.pdf.

65. AIDSinfo. Guidelines for the Use of Antiretroviral Agents in HIV- 1-Infected Adults and Adolescents.
Updated 2016 Jul. Available from: https://aidsinfo.nih.gov/guidelines/html/1/adult-andadolescent- arv-
guidelines/31/adverse-effects-of-arv

66. Maigetter K, Pollock AM, Kadam A, Ward K, Weiss MG. Pharmacovigilance in India, Uganda and South Africa
with reference to WHO’s minimum requirements. Int J Health Policy Manag. 2015 Mar 9;4(5):295-305.

67. Lemoine M, Nayagam S, Thursz M. Viral hepatitis in resourcelimited countries and access to antiviral
therapies: current and future challenges. Future Virol. 2013 Apr;8(4):371-380.

68. Franco E, Bagnato B, Marino MG, Meleleo C, Serino L, Zaratti L. Hepatitis B: Epidemiology and prevention
in developing countries. World J Hepatol. 2012 Mar 27;4(3):74-80.

69. Liu Z, Hou J. Hepatitis B virus (HBV) and hepatitis C virus (HCV) dual infection. Int J Med Sci. 2006;3(2):57-62.

70. FDA. FDA Drug Safety Communication: FDA warns about the risk of hepatitis B reactivating in some
patients treated with directacting antivirals for hepatitis C. 2016 Oct. Available from: https:// www.fda.gov/
Drugs/DrugSafety/ucm522932.htm

71. World Health Organization. Global Report on Access to Hepatitis C Treatment Focus on Overcoming
Barriers. Last updated 2016 Oct. Available from: http://apps.who.int/iris/ bitstream/10665/250625/1/
WHO-HIV-2016.20-eng.pdf?ua=1

48
72. World Health Organization. Globocan 2012: Estimated Cancer Incidence, Mortality, and Prevalence
Worldwide in 2012. Available from: http://globocan.iarc.fr/Pages/fact_sheets_cancer.aspx

73. Cancer research UK. Breast Cancer Surivival Statistics. 2014 Nov. Available from: http://www.
cancerresearchuk.org/healthprofessional/ cancer-statistics/statistics-by-cancer-type/breastcancer/
survival#heading-Zero

74. Vanderpuye V, Grover S, Hammad N, PoojaPrabhakar, Simonds H, Olopade F, Stefan DC. An update on the
management of breast cancer in Africa. Infect Agent Cancer. 2017 Feb 14;12:13.

75. Katanoda K, Matsuda T. Five-year relative survival rate of breast cancer in the USA, Europe and Japan. Jpn
J Clin Oncol. 2014 Jun;44(6):611.

76 OECD, New Health Technologies: Managing Access, Value and Sustainability, 2017. Available from:
http://www.oecd-ilibrary.org/ social-issues-migration-health/managing-new-technologies-inhealth-
care_9789264266438-en

77. BBC News. Google DeepMind’s NHS deal under scrutiny. 2017 Mar. Available from: http://www.bbc.com/
news/ technology-39301901

78. Dzau VJ, Ginsburg GS. Realizing the Full Potential of Precision Medicine in Health and Health Care. JAMA.
2016 Oct 25;316(16):1659-1660.

79. Schork NJ. Personalized medicine: Time for one-person trials. Nature. 2015 Apr 30;520(7549):609-11.

49
Footnotes
i. For example, in ALK positive NSCLC patients who previously were treated with chemotherapy, crizotinib
(Xalkori) demonstrated an overall response rate of (ORR) of 65% and a median progression free survival
rate of 7.7 months compared to chemotherapy, alone (20% and 3.0 months, respectively).3 In another
example, the use of two systems therapeutics, pertuzumab (Perjeta) and trastuzumab (Herceptin),
combined with the chemotherapy agent docetaxel, demonstrated an overall response rate (ORR) of 80.2%
for the treatment of HER2 positive breast cancer patients compared to an ORR of 69.3% in the same patient
population with trastuzumab and docetaxel. In the same trial, the median duration of response was nearly
double at 20.2 months versus 12.5 months.4

ii. For example, the prescribing information for paroxetine notes that paroxetine is metabolized by CYP2D6
and suggests to avoid co-prescribing with known CYP2D6 inhibitors, because and co-prescribing could
lead to adverse events such as prolonged QTC levels. However, the label does not specifically state
patients should be tested for pharmacokinetic profile prior to receiving the therapy. Genetic variations
of CYP2D6 are widespread, causing some individuals to be ultra-rapid, intermediate, or poor enzyme
metabolizers. CYP2D6 metabolizes a significant number of commonly used drug classes such as
antidepressants, antipsychotics, and opioids and so plays a significant role in drugdiscovery as well as
current treatment paradigms.

iii. Breast cancer is one of the leading cancers globally (1.7 million cases in 2012), and ER positive breast
cancers are a common variant (70–80%), which leads to high volume for breast cancer medicines.26

iv. Tamoxifen, anastrozole, and letrozole are all available as generic options and have a greater use in EU5 than in
the United States, which is reflected in this high volume of stratified oncology therapies in this region.

v. These medicines are a success story for personalized oncology as they helped to revolutionize the
treatment of patients with Philadelphia chromosome positive chronic myeloid leukemia (Ph+ CML). Over
90% of patients with typical CML have this genotype, and it is part of the diagnostic criteria to test for the
status of this variant. Another leukemia therapy, ibrutinib, is the only therapy among the top 10 highest
volume oncology products to have been launched within the last five years. Ibrutinib is an oral, Burton’s
tyrosine kinase inhibitor with regulatory approval across the developed countries for chronic lymphocytic
leukemia (CLL)/small lymphocytic lymphoma that (SLL) with 17p deletion. Ibrutinib is recommended for
use in high risk CLL (those patients harboring a 17p deletion). Historically, patients with the 17p deletion
genotype had poor outcomes with conventional oncology regimens.27

vi. In the United States in 2016, Harvoni’s list price was $94,500 per 12-week course of treatment and Sovaldi
was $84,000, although in practice most US payers received these therapies at a discounted gross price.29

vii. An analysis of Medicare reimbursement in the United States of ten drug-diagnostic combinations found
that although the therapies themselves were reimbursed, there was “limited and variable reimbursement
of companion diagnostics for all new patients being prescribed the companion drug.” 32

50
vii. In the EU5, although patients have access to companion diagnostics, the scope and timing of patient
access to diagnostics varies between countries; for example, funding of companion diagnostic testing is
mandatory if required by the drug label in Germany, while in in France, the National Institute of Cancer
(Institut National du Cancer; INCa) covers the cost of molecular testing of an associated oncology
medicine at the time of the corresponding drug launch but only on a temporary basis.33

ix. A 2015 statistical analysis assessed the impact of companion diagnostic measurement on treatment effects
and sample sizes for the clinical trials.36 The results support the idea that wrongly stratifying patients based
on inaccurate measurements of companion diagnostic tests leads to lower perceived efficacy in clinical
trials and that there is a direct relationship between dilution of efficacy in a clinical trial and increasing
inaccuracy of a companion diagnostic

x. Taking another example from oncology, although close to 100% of new cancer medicines were reimbursed
in the United States and Germany from 2014–2015, less than half of these medicines were reimbursed in
other countries, including the UK, Australia, Sweden, and Italy.22

xi. About half of cutaneous melanoma patients can be stratified by BRAF mutation to be eligible for treatment
with BRAF inhibitors such as vemurafenib, trametinib, or dabrafenib; monotherapy with these agent
has shown improved progression-free survival (PFS) and overall survival (OS) compared to traditional
chemotherapy, alone. However, disease progression can occur within 6 to 8 months due to development of
resistance mechanisms. Combination of a BRAF inhibitor with a MEK inhibitor (the current gold standard)
has shown benefits in PFS, but can still lead to resistance and each combination of therapies has slightly
different toxicity profiles.

xii. Regulatory bodies are responsible for post marketing safety monitoring for all medicines. According to
the world health organization, “while spontaneous reporting remains a cornerstone of pharmacovigilance
in the regulatory environment… the need for more active surveillance has also become increasingly clear.
Without information on utilization and on the extent of consumption, spontaneous reports do not make it
possible to determine the frequency of an adverse drug events attributable to a product, or its safety in
relation to a comparator. More systematic and robust epidemiological methods that take into account the
limitations of spontaneous reporting are required to address these important safety questions. They need
to be incorporated into post-marketing surveillance programs.”41

xiii. In developed countries, governments have taken initiatives to support precision medicine and genomic
initiatives, such as the United Kingdom’s 100,000 Genomes Project and the United States’ Precision Medicine
initiative; despite this, the right patients for a specific systems therapeutic may not receive testing.

xiv. In the United States, the FDA sets regulations for pharmacovigilance along with the divisions of the Center
for Drug Evaluation and Research and the Center for Biologics Evaluation and Research, within the FDA.
Post-marketing ADRs are reported to the FDA through MEDWatch, either voluntarily by stake holders or
through mandatory reporting, and are assessed by the divisions mentioned, previously.

51
Footnotes
xv. In United States, a 1994 study estimated 100,000 annual hospital deaths from adverse drug reactions
(ADRs); this number was subsequently extrapolated to the population of the European Union to account
for 197,000 annual hospital deaths from ADRs in 2006.

xvi. Known adverse events to HIV antiviral therapies include neutropenia, which is one of the most
predominant adverse events in Africa which suggests that patients are experience a high rate of ADRs that
can be attributed to antiretroviral therapies.64

xvii. For the cases reported, two patients died and another patient required a liver transplant. According to the
World Health Organization, 275,000 people living in low- and middle-income countries have received DAA
hepatitis C treatment in 2015.70 71

xviii. According to one study, the lack of support services to manage the toxicity and adverse events associated
with treatment can be a reason to withhold treatment even when it is available.74

52
Methodology
IDENTIFICATION OF STRATIFIED MEDICINES
To identify stratified medicines, we examined information on the PharmGKB (https://www.pharmgkb.org/), FDA
(Drugs@FDA and the FDA’s Table of Pharmacogenomic Biomarkers in Drug Labeling), European Medicines
Agency (EMA), Health Canada and Pharmaceuticals and Medical Devices Agency (PMDA), Japan, through 2016.
We included those therapies whose labels require or recommend testing; we excluded therapies where biomarker
information was included but was informational. In addition, we included therapies which were stratified according
to viral genotype, such as sofosbuvir for HCV-1, HCv-2, HCV-3 and HCV-4. Two of the therapies were included
that were outside this definition. Buserelin, which is available in Canada and Europe, is indicated for hormone
dependent prostate cancer and so hormone status testing is necessary prior to use. Tamoxifen does not have a
labeling requirement for hormonal testing prior to use, but guidelines suggest testing prior to use.

DATA ON UTILIZATION OF STRATIFIED MEDICINES


To assure consistent geographic and intertemporal measurement of stratified medicine utilization, we used the
IQVIA MIDAS database, which reports standard units (SUs) and dollar expenditures (evaluated at manufacturers’
prices using annual exchange rates) for marketed therapeutics in the ten developed countries. This database
accounts for approximately 97% of global therapeutic usage, and utilizes standardized definitions and collection
processes. For eight medicines having both biomarker requirements and indications that do not have biomarker
requirements prior to use — rituximab (Rituxan), nivolumab (Opdivo), ibrutinib (Imbruvica), erolotinib (Tarceva),
pembrolizumab (Keytruda), cetuximab (Erbitux), atezolizumab (Tecentriq), everolimus (Afinitor) — we allocated
stratified medicine sales and volume by indication using indication-specific analyses based on IQVIA anonymized
patient level data.

These methods have some limitations. For example, existing global data sources do not link diagnostic testing
to therapeutic use. Thus we were unable to determine whether personalized medicines are actually utilized in
accordance with their labelling and treatment guidelines. Although we were able to measure use of personalized
medicines reasonably accurately, we were not able to estimate appropriate use.

53
STRATIFIED MEDICINES

SAMPLE APPROVAL BIOMARKER THERAPEUTIC


MOLECULE BIOMARKER DX TEST PURPOSE
BRAND YEAR APPROVAL AREA

Adverse Drug
abacavir HLA-B Ziagen 1998 2008 Antiviral
Reaction

abacavir/ Adverse Drug


HLA-B Epzicom 2004 2008 Antiviral
lamivudine Reaction

abacavir/
Adverse Drug
lamivudine/ HLA-B Trizivir 2000 2008 Antiviral
Reaction
zidovudine

ado-trastuzumab
HER2+ Kadcyla 2013 2013 Oncology Efficacy
emtansine

Gilotrif
afatinib EGFR 2013 2013 Oncology Efficacy
GiIlotrif

alectinib ALK+ Alecensa 2015 2015 Oncology Efficacy

anastrozole ER+ Arimidex 1995 2000 Oncology Efficacy

t(15;17) translocation
arsenic trioxide and/or PML/RAR Trisenox 2000 2000 Oncology Efficacy
fusion gene

Adverse Drug
atazanavir CYP2C19 Reyataz 2003 n/a Antiviral
Reaction

atezolizumab PD-L1 Tecentriq 2016 2016 Oncology Efficacy

Adverse Drug
azathioprine TPMT Imuran 1968 2005 Immunology
Reaction

bosutinib BCR-ABL1 Bosulif 2012 2012 Oncology Efficacy

hormone-dependent
buserelin Suprefact 2003 2009 Oncology Efficacy
prostate cancer

Adverse Drug
carbamazepine HLA-B*1502 Tegretol 1968 2007 CNS
Reaction

CARBAGLU- Inborn Errors of


carglumic acid NAGS 2010 2010 Efficacy
Carbaglu Metabolism

ceritinib ALK Zykadia 2014 2014 Oncology Efficacy

cetuximab EGFR+/KRASwt Erbitux 2004 2006 Oncology Efficacy

AKR1D1, HSD3B7,
Inborn Errors of
cholic acid CYP27A1, AMACR, Cholbam 2015 2015 Efficacy
Metabolism
CYP7A1

cobimetinib BRAF Cotellic 2015 2015 Oncology Efficacy

crizotinib ALK/ROS1 Xalkori 2011 2015 Oncology Efficacy

dabrafenib BRAF Tafinlar 2013 2013 Oncology Efficacy

daclatasvir HCV 3 Daklinza 2015 2015 Antiviral Efficacy

54
STRATIFIED MEDICINES

SAMPLE APPROVAL BIOMARKER THERAPEUTIC


MOLECULE BIOMARKER DX TEST PURPOSE
BRAND YEAR APPROVAL AREA

BCR-ABL1
(Philadelphia
dasatinib Sprycel 2006 2006 Oncology Efficacy
chromosome-positive
(Ph+))

denileukin diftitox CD25+ Ontak 1999 1999 Oncology Efficacy

dextromethorphan/ Adverse Drug


CYP2D6 Nuedexta 2010 2010 CNS
quinidine Reaction

Adverse Drug
divalproex sodium POLG Depakote 1983 n/a CNS
Reaction

elbasvir/grazoprevir HCV 1, 4 Zepatier 2016 2016 Antiviral Efficacy

Inborn Errors of Dosing and


eliglustat CYP2D6 Cerdelga 2014 2014
Metabolism Efficacy

Inborn Errors of
elosulfase alfa GALNS- Vimizim 2014 2014 Efficacy
Metabolism

erlotinib EGFR Tarceva 2004 2016 Oncology Efficacy

gene that is amenable


eteplirsen Exondys 51 2016 2016 Genetic Disorder Efficacy
to exon 51 skipping

everolimus ER+, HER2- Afinitor 2009 2012 Oncology Efficacy

exemestane ER+ Aromasin 1999 2005 Oncology Efficacy

fulvestrant HR+ Faslodex 2002 2002 Oncology Efficacy

EGFR-TK activating
gefitinib Iressa 2003 2009 Oncology Efficacy
mutations

gemtuzumab
CD33+ Mylotarg 2000 2000 Oncology Efficacy
ozogamicin

ibrutinib del (17p) Imbruvica 2013 2014 Oncology Efficacy

c-Kit+ or CD117+(Kit),
Dosing and
imatinib BCRABL1, PDGFRB, Gleevec 2001 2002 Oncology
Efficacy
FIP1L1-PDGFRA

ivacaftor CFTR Kalydeco 2012 2013 Respiratory Efficacy

ivacaftor /
CFTR Orkambi 2015 2015 Respiratory Efficacy
lumacaftor

lapatinib HER2+ Tykerb 2007 2012 Oncology Efficacy

ledipasvir/
HCV 1,4, 5,6 Harvoni 2014 2014 Antiviral Efficacy
sofosbuvir

lenalidomide 5q deletion Revlimid 2005 2005 Immunology Efficacy

letrozole ER+ Femara 1997 2001 Oncology Efficacy

maraviroc CCR5 tropic HIV-1 Selzentry 2007 2007 Antiviral Efficacy

55
STRATIFIED MEDICINES

SAMPLE APPROVAL BIOMARKER THERAPEUTIC


MOLECULE BIOMARKER DX TEST PURPOSE
BRAND YEAR APPROVAL AREA

Dosing, Efficacy
mercaptopurine TPMT Purinethol 1953 2004 Oncology and Adverse Drug
Reaction

BCR-ABL
Efficacy and
(Philadelphia
nilotinib Tasigna 2007 2007 Oncology Adverse Drug
chromosome-positive
Reaction
(Ph+)); UGT1A1

nivolumab BRAF Opdivo 2014 2014 Oncology Efficacy

olaparib BRCA1-2 Lynparza 2014 2014 Oncology Efficacy

ombitasvir/
paritapre-vir/ HCV 4 Technivie 2015 2015 Antiviral Efficacy
ritonavir

ombitasvir/
paritaprevir/
HCV 1 Viekira Pak 2014 2014 Antiviral Efficacy
ritonavir/dasabuvir
sodium

osimertinib EGFR Tagrisso 2014 2016 Oncology Efficacy

oxcarbazepine
HLA-B*1502 Oxtellar XR 1999 2006 CNS Adverse Drug
Reaction

palbociclib ESR1, ERBB2 Ibrance 2015 2015 Oncology Efficacy

panitumumab EGFR+, KRASwt Vectibix 2006 2006 Oncology Efficacy

pegloticase
G6PD Krystexxa 2010 2010 Rheumatology Adverse Drug
Reaction

pembrolizumab CD274/PD-L1 Keytruda 2014 2015 Oncology Efficacy

pertuzumab ERBB2 Perjeta 2012 2012 Oncology Efficacy

deficiencies in CPS, BUPHENYL- Inborn Errors of


phenylbutyrate 1996 2009
OTC, or AS Buphenyl Metabolism

Dosing and
phenytoin CYP2C9,HLA-B Dilantin 1953 2008 CNS Adverse Drug
Reaction

pimozide CYP2D6 Orap 1984 2011 CNS Dosing

ponatinib BCR-ABL1 Iclusig 2012 2012 Oncology Efficacy

rasburicase
G6PD deficiency Elitek/ 2002 2002 Rheumatology Adverse Drug
Reaction

Oncology &
rituximab CD20+subset Rituxan 1997 1997 Efficacy
Immunology

rucaparib BRCA Rubraca 2016 2016 Oncology Efficacy

56
STRATIFIED MEDICINES

SAMPLE APPROVAL BIOMARKER THERAPEUTIC


MOLECULE BIOMARKER DX TEST PURPOSE
BRAND YEAR APPROVAL AREA

simeprevir HCV 1 Olysio 2013 2013 Antiviral Efficacy

sodium
NAGS, CPS1, OTC, Efficacy of
phenylacetate and Ammonul 2005 2005 Inborn Errors
ASS1, ASL and ARG1 Metabolism
sodium benzoate

Solvadi/
sofosbuvir HCV 1,2,3,4 2013 2013 Antiviral Efficacy
Sovaldi

sofosbuvir/
HCV 1,2,3,4,5,6 Epclusa 2016 2016 Antiviral Efficacy
velpatasvir

tamoxifen ER+ Nolvadex 1977 1977 Oncology Efficacy

XENAZINE
tetrabenazine CYP2D6 Xenazine, 1950 2008 CNS Dosing
Nitoman

toremifene ER+ Fareston 1997 1997 Oncology Efficacy

tositumomab
CD20+ Bexxar 2003 2003 Oncology Efficacy
iodine-131

trametinib BRAF Mekinist 2013 2013 Oncology Efficacy

trastuzumab HER2 + Herceptin 1998 2004 Oncology Efficacy

t(15;17) translocation
tretinoin and/ or PML/RAR Vesanoid 1995 1995 Oncology Efficacy
fusion gene

vandetanib RET Caprelsa 2011 2011 Oncology Efficacy

Inborn Errors of
velaglucerase alfa GBA Vpriv 2010 2010 Efficacy
Metabolism

vemurafenib BRAF/NRAS Zelboraf 2011 2014 Oncology Efficacy

venetoclax 17p deletion Venclexta 2016 2016 Oncology Efficacy


57
About the authors
MURRAY AITKEN ALANA SIMORELLIS, PH.D.
Executive Director, IQVIA Institute Publications Manager, IQVIA
for Human Data Science Institute for Human Data Science

Murray Aitken is a Senior Vice Alana is Publications Manager for


President at IQVIA and the executive the IQVIA Institute for Human Data
director of the IQVIA Institute for Human Data Science. Science and helps manage aspects of IQVIA Institute for
Murray is a renowned healthcare expert on addressing Human Data Science research projects and publications
the challenges facing the global healthcare industry as well as conducting research and analysis within
and prospects for improving patient outcomes, global healthcare. Alana came to IQVIA in 2016 having
managing costs and maximizing access through better previously worked at Decision Resources Group for
use of healthcare data and information. Established over six years as a Principal Business Insights Analyst.
in 2011, the IQVIA Institute for Human Data Science At Decision Resources group, Alana authored a number
provides global policy setters and decision makers of publications within multiple disease areas that
with objective, transformational insights into healthcare included Alzheimer’s disease, pain, bipolar disorder,
dynamics derived from granular analysis of information. schizophrenia and major depression. Alana has a Ph.D.
in Chemistry from the University of Utah and completed
a post-doctoral fellowship at Brandeis University, where
part of her research involved structural investigation of
a protein associated with Parkinson’s disease.

58
About the Institute
The IQVIA Institute for Human Data Science contributes • Understanding the future role for biopharmaceuticals
to the advancement of human health globally through in human health, market dynamics, and implications
timely research, insightful analysis and scientific for manufacturers, public and private payers,
expertise applied to granular non-identified patient- providers, patients, pharmacists and distributors.
level data.
• Researching the role of technology in health system
Fulfilling an essential need within healthcare, the products, processes and delivery systems and the
Institute delivers objective, relevant insights and business and policy systems that drive innovation.
research that accelerate understanding and innovation
critical to sound decision making and improved Guiding Principles
human outcomes. With access to IQVIA’s institutional The Institute operates from a set of Guiding Principles:
knowledge, advanced analytics, technology and
unparalleled data the Institute works in tandem with a • Healthcare solutions of the future require fact based
broad set of healthcare stakeholders to drive a research scientific evidence, expert analysis of information,
agenda focused on Human Data Science including, technology, ingenuity and a focus on individuals.
including government agencies, academic institutions,
• Rigorous analysis must be applied to vast amounts of
the life sciences industry and payers.
timely, high quality and relevant data to provide value
Research Agenda and move healthcare forward.
The research agenda for the Institute centers on 5 areas
• Collaboration across all stakeholders in the
considered vital to contributing to the advancement of
public and private sectors is critical to advancing
human health globally:
healthcare solutions.

• Improving decision-making across health systems • Insights gained from information and analysis should
through the effective use of advanced analytics and be made widely available to healthcare stakeholders.
methodologies applied to timely, relevant data.
• Protecting individual privacy is essential, so research will
be based on the use of non-identified patient information
• Addressing opportunities to improve clinical
and provider information will be aggregated.
development productivity focused on innovative
treatments that advance healthcare globally. • Information will be used responsibly to advance
research, inform discourse, achieve better healthcare
• Optimizing the performance of health systems by and improve the health of all people.
focusing on patient centricity, precision medicine
and better understanding disease causes, treatment
consequences and measures to improve quality and
cost of healthcare delivered to patients.

59
CONTACT US
iqvia.com/contactus

LOCATION
100 IMS Drive
Parsippany, NJ 07054
USA

Copyright © 2017 IQVIA. All rights reserved. PB.0007-1-11.2017

Das könnte Ihnen auch gefallen