Sie sind auf Seite 1von 56

Pharma R&D Compendium i

2009

Invention
reinvented
McKinsey perspectives on pharmaceutical R&D
Pharma R&D Compendium 2010 i

Invention
reinvented
McKinsey perspectives on pharmaceutical R&D
2010

Head of Global and North American Pharmaceutical and Medical Products R&D Practice
Rodney Zemmel

Head of European Pharmaceutical and Medical Products R&D Practice


Mubasher Sheikh

For more information please contact


pharma_r&d@mckinsey.com

London New Jersey New York


No. 1 Jermyn Street 600 Campus Drive 55 East 52nd Street
London Florham Park 21st Floor
SW1Y 4UH NJ-07932-1046 New York, NY-10022
United Kingdom United States United States
Tel: +44 (20) 7839 8040 Tel: +1 (973) 549 6600 Tel: +1 (212) 446 7000

Silicon Valley Zurich


3075A Hansen Way Hotelstraße
Palo Alto Zürich-Flughafen
CA-94304 CH-8060
United States Switzerland
Tel: +1 (650) 494 6262 Tel: +41 (44) 876 8000
ii Pharma R&D Compendium 2010 iii

Contents

1. The road to positive R&D returns 51. Changing with the times: how pharmaceutical companies should
Scientific innovation is not the only way to higher R&D productivity. respond to regulatory change
Attention to the familiar management areas of cost, speed, and decision Drug companies need to make organizational changes to meet the latest
making can still reap rewards. patient safety regulations in the United States. But as the regulations
Eric David, Tony Tramontin, and Rodney Zemmel evolve, companies will also need to work openly with all stakeholders to
ensure the right outcome.
Maria Gordian and Richa Pande
7. The secret of high productivity in the research lab
What drives research productivity? An understanding of how the world’s
most successful laboratories operate reveals some answers. 57. The anatomy of attrition
Mark Beards, Michael Edwards, and Mubasher Sheikh Research tracking the attrition rates of more than 3,000 compounds in
each phase of development reveals recent drug failure trends.
Navjot Singh, Rodney Zemmel, Maria Gordian, and Áine Denari
15. The future of drug development: advancing clinical trial design
Traditionally, drug development has been conducted in distinct—and
increasingly expensive—phases. A more integrated model that uses 65. What gives drugs the X factor?
adaptive design approaches to enhance flexibility and maximize the use of Despite the long development process, more than two-thirds of new drugs
accumulated knowledge could improve the quality and reduce the cost. launched on the market are commercial failures. Recent research uncovers
John Orloff, Frank Douglas, Jose Pinheiro, Susan Levinson, what distinguishes those that succeed.
Michael Branson, Pravin Chaturvedi, Ene Ette, Paul Gallo, Gigi Hirsch, Ramesh Hariharan and Navjot Singh
Cyrus Mehta, Nitin Patel, Sameer Sabir, Stacy Springs, Donald Stanski,
Matthias Evers, Edd Fleming, Navjot Singh, Tony Tramontin, and 73. A special kind of success
Howard Golub Mainstream pharmaceutical companies are turning their attention to
specialty drugs. Which factors are most important for commercial success
or failure?
33. Using IT to speed up clinical trials Mark Gudiksen, Edd Fleming, Laura Furstenthal, and Philip Ma
The first wave of IT improvements solved some problems, but others
remain. Now it is time for a more comprehensive approach.
Sam Marwaha, Samir Patil, and Navjot Singh 81. R&D in emerging markets: a new approach for a new era
Gone is the time when R&D in developed markets could meet the
pharmaceutical needs of emerging markets. Local presence is required.
41. Quality gates: raising the standard of Michael Edwards
pharmaceutical technical development
Good technical development can improve quality, lower costs, and get
products to market more quickly, yet few pharmaceutical companies 87. The microeconomics of personalized medicine
manage it well. Quality gates can transform the process. Personalized medicine promises to increase the quality of clinical care
Michele Holcomb, Martin Møller, and Paul Rutten and, in some cases, decrease health care costs. The biggest hurdles are
economic, not scientific.
Jerel Davis, Laura Furstenthal, Amar Desai, Troy Norris, Saumya Sutaria,
47. Market-access-minded development Edd Fleming, and Philip Ma
A new way of conceiving clinical development will uncover the drugs
bound for commercial success early on. Think value.
Matthias Evers 101. Authors’ contact details
iv Pharma R&D Compendium 2010 v

Invention reinvented
Introduction
In the eyes of patients and other stakeholders, the pharmaceutical industry exists
to discover new medicines that go on to become standard treatments. The faltering
economics of R&D productivity are jeopardizing that mission: R&D expenditure is
not delivering.

According to analysis, costs have ballooned to more than $1.6 billion per new drug
approved, compared with $1 billion in 2005, attrition rates remain stubbornly high, and
many of the drugs that do eventually reach the market fail to gain broad acceptance.
Today, only 30 percent of drugs launched earn an acceptable rate of return—a rate
that is likely to deteriorate further given the increasing demands of payors and access
agencies that want more value for their money. In 2008, for example, 96 percent of
drugs reviewed by the French authorities were classified as having “minor differentiation
at best,” meaning limited market access. And between 1998 and 2008 in the United
Kingdom, almost 60 percent of drugs from the top ten pharmaceutical companies were
given negative or restricted access. The recent global economic crisis can only make
payors even more sensitive to costs.

Many have long called for what is effectively a new R&D paradigm—although
apparently to little effect, given that drug approvals continue to fall. Today however, we
sense real change as companies learn to adapt to a far harsher environment. Invention
is being reinvented.

In this collection of articles, we set out our thoughts on some of the elements of
that reinvention. We examine the untapped scope for improving productivity not
through scientific innovation, but through better management of R&D and the use
of IT in clinical trials. We publish original research on why so many drugs fall out of
the development pipeline, and on what makes some laboratories more productive
than others. We analyze the factors that account for commercial success, and look
at how pharmaceutical companies are changing the way they work and think to
meet new regulatory safety requirements and to gain market access. We assert
that clinical development can be improved by moving from the traditional sequential
approach to a more integrated model. And we investigate emerging opportunities—
dissecting the economics of personalized medicine, and explaining why now is
the time to refocus R&D on the needs and preferences of developing markets.

We hope you will find these ideas stimulating and welcome your comments. Please
send them to pharma_r&d@mckinsey.com, or contact any of the authors individually.

Rodney Zemmel Mubasher Sheikh


Director, New York office Principal, London office
Pharma R&D Compendium 2010 1
The road to positive R&D returns

Scientific innovation is not the only way to higher R&D


The road to positive R&D returns productivity. Attention to the familiar management areas
of cost, speed, and decision making can still reap rewards.

Eric David, Tony Tramontin, and Rodney Zemmel

As productivity in the pharmaceuticals Sizing the challenge


industry has fallen, so calls for a “new
paradigm” that will radically change We modeled the estimated average
pharmaceutical R&D have increased return on R&D investment for a typical
(Exhibit 1). The trend has been to small-molecule compound and for a
view diminishing returns as a science typical biologic. With net present value
problem. But while scientific innovation (NPV) of minus $65 million and an IRR of
is certainly part of the solution, 7.5 percent, the model suggested that
management should not overlook present-day returns for an average small
other, more familiar means of value molecule fall below the cost of capital
creation. Increased attention to costs, (Exhibit 3). By contrast, between 1997 and
speed of development, and decision 2001, the return approached 12 percent.
making could increase the internal
rate of return (IRR) of an average small The key factors driving this downward
molecule from around 7.5 percent— trend in productivity are well known.
which is less than the industry’s cost Industry interviews and analysis of the
of capital—to 13 percent (Exhibit 2). Pharmaprojects database indicate that,
over the past decade,
the overall probability of
EXHIBIT 1 success (POS) for small
More talk, fewer approvals molecules has fallen by
FDA NME approvals
five percentage points,
1

PubMed occurrences of

40
“new pharmaceutical and the time required
research paradigm”2

for R&D has increased


35
by between 12 and 18
30
months. Furthermore, R&D
25
costs have recently risen by
20
about 8 percent annually,
15
while prices worldwide
10
are under pressure.
5

0
1998 2003 2008
It could be argued that
companies should shift
1
New molecular entity.
much of their R&D
investment to biologics,
2
PubMed search for “new pharmaceutical research paradigm.” The term “cardiac surgery” was used as a control over the same
time period to ensure trends were not simply due to changes in the number of publications available in PubMed over time.
Source: NME data from Parexel’s Pharmaceutical R&D Statistical Sourcebook 2008/2009
2 Pharma R&D Compendium 2010 3
The road to positive R&D returns

as the average biologic EXHIBIT 2 EXHIBIT 4 program by a conservative


currently offers a greater More value from R&D Slim returns on small molecules 18 months. This increased
Successfully launched small molecules, IRR by sales quartile,1 %
return (NPV of $1.26 1.0%
the NPV of an average
13.0%
billion, IRR of 13 percent) 1.5%
1.0%
28 compound by about
owing to higher average 2.0%
$190 million, raising the
7.5%
peak sales and slower IRR by 1.5 percentage
decay of sales following points. Some companies
12
loss of exclusivity. But IRR =
WACC =
2
8
have done much better:
given the limited number Current Cost Speed Decision Decision New IRR for
9.5% 6
Merck accelerated the
IRR for 1
making 1 making 2 average small
of such molecules and average
small
moecule launch of the diabetes
the expected erosion of molecule First
quartile
Second
quartile
Third
quartile
Fourth
quartile
drug sitagliptin (Januvia)
returns as competition by three to four years
Reduce Reduce Increase Shift 4% of
from biosimilars mounts, overall cost
per molecule
time-to-
launch by
Phase III
POS 10%
molecules
from fourth 2
Percentage of
drugs in each
2 4 40 54 by employing novel
increased investment in by 15% 18 months by “taking”
attrition in
quartile to
first quartile
sales quartile
1
parallel development
Sales by quartile are based on a historic sample (2000 to 2006) of successfully launched drugs; NPV is calculated assuming
biologics alone is not the Phase II
0% contribution margin for first 2 years, 50% margin for years 3-4, 70% for years 6-10, and 60% thereafter; 9.5% discount
rate. Only R&D costs directly associated with launched molecules are reflected in NPV (cost of attrition accounted for
techniques. But gains
solution. Rather, more Internal rate of return.
1

Probability of success.
2 2
elsewhere).
Weighted average cost of capital.
in speed cannot come
careful management from short cuts: the key
attention to reducing to capturing value from
costs and accelerating time-to-launch, aggressive outsourcing of selected non- Lilly’s Chorus unit represents one effort to program acceleration is choosing
and better decision making over when core activities to low-cost geographies. reduce costs by focusing on the activities the right programs to accelerate.
to remove poorly performing drugs from A second approach is to reduce the costs that truly reduce the risk of failure of a
the portfolio and which compounds to associated with drug failures. Companies compound on the way to proof of concept. Decision making
invest in, will reap significant rewards. generally design R&D programs for The cost of failure can also be lessened R&D leaders grapple with decisions
success, even though the majority by sharing risk with another party, such about program termination, acceleration,
Levers for change of programs will fail. Costly, two-year as another pharmaceutical company, resourcing, and prioritization. Project-
carcinogenicity studies, for example, are a contract research organization, or termination decisions are especially
Costs often initiated before a compound reaches investors. Such strategies can together difficult and can cost a company
Although most companies have made proof of concept at the end of Phase reduce the overall cost of R&D by 15 hundreds of millions of dollars if made
progress in reducing costs, efforts too IIa, and this expenditure is wasted if the percent or more, increasing the NPV too late. The current high attrition rate in
often focus on the obvious organizational compound fails (as it is likely to do). Eli of average small-molecule projects by Phase III trials suggests that companies
and procurement issues. about $250 million and raising the IRR of have overlooked or ignored key signals,
Successful companies EXHIBIT 3 R&D by some two percentage points. and in some cases made poor decisions
generally employ Falling below the cost of capital about aspects over which they have
$ million
broader strategies. R&D cost
Speed substantial control. Our analysis indicates
Small
molecules capitalized
at 9.5%1
1,097
Estimated NPV For medicines that make it to market that of 106 reported Phase III failures
(historic 272 294 318 343 370
236 for an average
One approach is for estimates
for average
0 0 96 131
small molecule successfully, our modeling indicates from 1990 to 2007, 45 percent were
is -$65 million,
companies to change what drug)
0% 50% 70% Year 10 value with an IRR of that each six-month delay to launch can due to insufficient efficacy of a drug
contribution contribution contribution of post-LOE 7.5%
they are doing, not just how -1,751
margin margin margin contributions3 mean a loss of almost $100 million in versus a placebo, and 24 percent to
they do it. For example, 2,691 NPV, or a reduction of 0.5 percentage insufficient differentiation versus standard
companies that consistently Biologics
R&D cost
points in IRR. This number is obviously of care. It is easy to scrutinize decision
capitalized
over-power clinical trials at 9.5%1
445 492 544 602 665 Estimated NPV much higher for top-performing drugs. making with the benefit of hindsight,
207 376 for an average
147
could reduce the number 0 0 biologic is
$1.26 billion,
Yet opportunities exist to address but R&D leaders can increase returns
of patients per trial. Our 0% 45% 65% Year 10 value
with an IRR of
13% inefficiencies such as poor planning by identifying and removing poor
contribution contribution contribution of post-LOE
experience also suggests margin2 margin2 margin2 contributions3 of clinical development, slow patient performers from the portfolio earlier in
-2,267
that R&D costs could be 1
Fully loaded, attrition-adjusted R&D costs are capitalized to their future value in the year of the drug’s launch and treated as
recruitment, and suboptimal site and development. Many organizations still
reduced by between 5 and 2
one-time investment in the IRR calculation.
Margins on biologics are assumed to be 5% lower than on small molecules to account for higher manufacturing costs.
investigator management. We modeled advance compounds for the wrong
10 percent through more the effect of accelerating a development reasons: because of “numbers-focused”
3
Year 10 value of post-LOE (loss of exclusivity) contributions discounted to year 10 at IRR; contributions derived from
sales assuming 60% margin for small molecules, and 55% for biologics.
Source: All new products since 2000 from EvaluatePharma; interviews; McKinsey analysis
4 Pharma R&D Compendium 2010 5
The road to positive R&D returns

incentive systems, because they fail to top-quartile drugs consistently: our


understand how product differentiation analysis shows that, in any given portfolio
is increasingly driving reimbursement, of small molecules, only 2 percent of
or because they have traveled too drugs will be top-quartile sellers, while
far down the development path. 54 percent will be fourth-quartile sellers.
However, by shifting even 4 percent of
Many companies have started to compounds from the fourth quartile to
restructure to address these issues. Eli the top quartile, the average IRR would
Lilly’s Chorus unit, GlaxoSmithKline’s increase by one percentage point.
Discovery Performance Units, and
Pfizer’s smaller, more focused therapeutic Implications for R&D leaders
areas are a few examples. If these
efforts enable R&D leaders to make A consistent, aggressive, and
better decisions and shift compound simultaneous focus on costs, speed, and
attrition to earlier stages, the impact decision making can raise the IRR on an
will be substantial. “Taking” attrition average small molecule from 7.5 percent
earlier during Phase II could increase to about 13 percent. For a typical portfolio
Phase III survival by ten percentage of a leading pharmaceutical company,
points—comparable to survival rates assuming a composition of 75 percent
between 1997 and 2001—and increase small molecules and 25 percent biologics
IRR by up to one percentage point. distributed across various phases of
development, this would raise the portfolio
Another key aspect of R&D decision return to between 14 and 15 percent,
making is the choice of compounds from between 9 and 10 percent currently.
in which to invest. We used data for
successfully launched drugs between Previous industry experience suggests
2000 and 2006 to divide products into such goals are attainable: from 1997
quartiles based on their returns. As to 2001, the return on the portfolio
shown in Exhibit 4, the top two quartiles described above would also have been
of launched molecules account for most 14 to 15 percent, driven by a higher
of the value creation. A top-quartile POS and shorter development times.
small molecule has an IRR of 28 percent Although the current environment
compared with the 7.5 percent of an is tougher, managers are not yet
average small molecule. A top-quartile fully exploiting the value-creation
biologic has an IRR of 33 percent levers described here, and moderate
compared with 13 percent for an average improvements can substantially increase
biologic. Second-quartile molecules returns. An IRR of 14 to 15 percent
have an IRR of 12 percent for a small on R&D might not sound like hitting
molecule and 15 percent for a biologic. the jackpot, but over a large portfolio
No company is likely to be able to identify it would create considerable value.

A version of this article, entitled “Pharmaceutical R&D: the road to positive returns,” was first
published in Nature Reviews Drug Discovery, August 2009, Volume 8, pp. 609–10.
For further details of the analysis and methodology, please contact the authors.

Eric David (eric_david@mckinsey.com) and Tony Tramontin (tony_tramontin@mckinsey.com) are


associate principals in McKinsey’s New York office, where Rodney Zemmel
(rodney_zemmel@mckinsey.com) is a director.
Pharma R&D Compendium 2010 7
The secret of high productivity in the research lab

What drives research productivity? An understanding of how the world’s


The secret of high productivity most successful laboratories operate reveals some answers.
in the research lab
Mark Beards, Michael Edwards, and Mubasher Sheikh

Increasing productivity is one of the different levels of performance, and


fundamental challenges of life sciences several more academic laboratories. The
research. The work is complex, lengthy, aim was to compare these laboratories’
and costly, and the failure rate high. practices with those identified in
But successful research can lead the first phase of the research (see
to disease-beating medicines and sidebar, “Research participants”).
significant financial rewards for the
research organizations involved. In the As a result, we identified many similarities
past, companies have tried to improve in the approaches adopted by all the
productivity through extensive, top- most productive laboratories, or “top
down initiatives, such as reorganizing labs.” Although the top labs were often
research or investing heavily in new conducting quite different kinds of
technology platforms. McKinsey, research, elements of their approach
through its “SuccessLab” initiative, in five areas—strategy decisions,
has now taken a different approach, talent management, portfolio and
setting out to understand what drives project management, problem solving,
research productivity from the bottom and collaboration—were remarkably
up—from the laboratory itself. similar. Be they in academia, the
pharmaceutical industry, high technology,
To grasp how the world’s leading or industrial chemical manufacturing,
research laboratories organize and top labs organize themselves to ensure
manage their research, we interviewed 12 they have the right team working
world-class academic innovators, including with a clear focus on a shared set
Nobel prize winners, McArthur Genius of activities, and that researchers
Award winners, and some of the world’s spend as little time as possible on
most highly cited scientists.1 We wanted administration and management.
to understand what might differentiate The result is higher productivity.
their approach from that used in other,
less productive laboratories, and whether Although there are structural differences
there were key practices and processes in how the various top labs try to achieve
that accounted for their success. A clear this outcome, the underlying principles
pattern emerged from the interviews. are often consistent. Below we describe
these principles, which together, our
We then examined the practices of 15 research suggests, amount to a best-
research laboratories in industry with practice approach to higher productivity.
8 Pharma R&D Compendium 2010 9
The secret of high productivity in the research lab

Research participants effective decision making on allocation of and reputation through working with turnover, although this can be more
resources, the building of new capabilities, academic institutions, offering PhD difficult to achieve in industry than
12 Academic labs
and communication of a lab’s role among sponsorship and post-doctoral posts, academia. Seconding team members
• Dr Shizuo Akira, Osaka University
its own staff, throughout the wider presenting at conferences, taking to different parts of the organization
• Prof. Carolyn Bertozzi, UC Berkeley
• Prof. Sam Danishefsky, organization, and externally. The constant active roles in industry groups, and and exposing them to the different
Sloan-Kettering Cancer Center and search for ways to create competitive publishing. (The fact that they have a areas of focus within the laboratory’s
Columbia University advantage through emerging technologies clear strategic focus makes reputation portfolio can help to keep teams fresh.
• Prof. Ulrich Hartl, Max Planck Institute was another recurring strategic feature. building an easier task.) Internally,
of Biochemistry too, they work to attract talented Project and
• Sir Tim Hunt, Cancer Research UK Talent management individuals by way of secondments and portfolio management
• Prof. Steven Jackson, transfers, particularly in industry. Internal
University of Cambridge In evaluating researchers, top labs rate publications, poster sessions, and The top labs design their portfolios
• Prof. David Liu, Harvard University
intrinsic intellectual capability, scientific meetings serve to bring the laboratory to of projects to be interlinked, so that
• Prof. Eiichi Nakamura,
curiosity, and general problem-solving the attention of the wider organization. they are both additive, in that the lab
University of Tokyo
skills higher than specific technical reaps benefits from their intellectual
• Prof. Svante Pääbo,
Max Planck Institute for Evolutionary knowledge. There are of course occasions Once they are in place, new recruits scale, and synergistic, in that each
Anthropology when a specific technical capability is are given formal and informal support project might uncover insights that
• Dr Luca Scorrano, required. But even in these situations, and advice to help them to adopt the prove valuable to another. Competitive
University of Geneva and Venetian top labs will try to hire people with the laboratory’s high standards and practices advantage arises in both respects.
Institute of Molecular Medicine ability to adapt to new roles as the focus and to become useful as quickly as
• Prof. George Whitesides, of the lab evolves. To bring a range of possible. Existing team members are Project teams are assembled to
Harvard University perspectives and approaches to their expected to commit significant time to incorporate the mix of skills needed to
• Sir Greg Winter, MRC Laboratory of work they recruit widely, from different the one-on-one apprenticeship of new address particular problems, and it is
Molecular Biology
countries and institutions and from diverse joiners (their end-of-year evaluations these, rather than functional groups,
academic and commercial backgrounds. take account of this), and to assist in that are the focus of the organization.
12 Pharmaceutical labs, covering:
the continuous mentoring of junior lab Individual members too are expected to
• Discovery
• Medicinal chemistry One laboratory head believes that members. Encouraged in this way, new have expertise in several disciplines—the
• Technology “regular interviews are totally useless” hires and juniors feel their careers are same person might run biophysical
• Structural sciences as a way of assessing candidates and developing and that they are helped to and cell biological assays on p53,
• Lead evaluation prefers to rely on recommendations from reach their full potential. For a laboratory for example—rather than depth of
collaborators and former colleagues or to to do otherwise is to risk losing them. functional skills in one area, enabling
3 Additional labs ask applicants to work in the laboratory for the team to work flexibly to address
• Consumer electronics a trial period. Other recruiters supplement Financial incentives are transparently linked its needs at any point. Subject to the
• Semi-conductors interviews and technical assessments to certain achievements—an exceptional laboratory’s requirements, researchers
• Polymers
with problem-solving sessions, perhaps piece of research, or publication in a are allowed a degree of freedom to
asking candidates to spend a morning leading journal. At the other end of choose which projects they work on, and
The research project is continuing and the
devising answers to a specific question or the scale, poor performance is not staffing allocation is discussed with all
database expanding. To date, more than
1,000 scientists have participated. working in the laboratory with the team. tolerated for long, and researchers are researchers before decisions are made.
The latter serves the dual purpose of asked to leave if they do not respond
indicating how well the candidate will fit to efforts to improve their attainment. Once a team is assembled, members
in. With this in mind, some laboratories set about their project with a clear plan
Strategic decisions arrange a social occasion before hiring, Despite the efforts they make to build that details what should be achieved
and the views of existing team members the right teams, top labs are not overly by when and the mechanism for
Clear, three- to five-year strategies, are always sought before a decision is concerned about staff turnover, seeing reviewing progress. The plan is used
focused on portfolios of interconnected made. Aware that courtship is a two- it as an important way of preventing to forecast when particular resources
projects, were a shared characteristic of way street, top labs seek to attract the their problem-solving culture from or equipment might be required and
the top labs. Clarity of strategy enables best candidates by nurturing their profile stagnating. Some even encourage to anticipate potential bottlenecks or
10 Pharma R&D Compendium 2010 11
The secret of high productivity in the research lab

delays. Simple visual tools are used innovative ideas. Instead, they work with screened and six chosen to be presented you want things to spread as much as
to share information in a way that is the broad group to outline the general at the next club meeting. Each club possible like infectious diseases. You
quickly understood: for example, what approach. Their role is to ensure that member presents once every two to need crowdedness,” is the way one
is happening and when, or any likely the projects continue to contribute three weeks. The laboratory head says head of an academic laboratory puts it.
clashes in demand for equipment. to the overall goal of the laboratory he sees the club as a way of keeping
and are synergistic, not to micro everyone informed about what’s going Differences
But if all does not go according to plan, manage unless absolutely necessary. on in the wider world, as well as better between laboratories
if milestones are missed, top labs do Laboratory heads also encourage informed about what not to do.
not shy away from tough decisions. their researchers to spend time on Understanding what top labs do to
Although a plan might sometimes warrant projects driven by their own personal Generally, external collaboration with succeed makes it relatively easy to see
modification, they are keen to terminate interest. This could be defined as a other academic and industry laboratories why others that fall short of best practice
sooner rather than later any project that percentage of their time or as a set is not wide spread, and might be are less productive. If, for example, a
is not showing results. Pouring more period each week: one laboratory sets instigated only to address a specific laboratory has only a short-term strategy,
resources into a project that is unlikely aside Friday afternoons for work on problem. The reason is the perceived it is likely to struggle to identify where to
to add value, or allowing it to continue at personal initiatives. Allowing researchers need to protect a laboratory’s innovations. build capabilities, to link projects to get the
a lower level—because researchers are this freedom helps maintain their However, external collaboration can benefits of shared learning, and to articulate
reluctant to let go or because they feel passion for work, but can also ensure be a valuable source of ideas given the what the laboratory should be known for.
they might have missed something—robs innovative ideas are not overlooked. correct agreement structure, and top labs If it fails to recruit researchers according to
other projects that have greater potential. Some laboratories have seen their see it as an important element of their best-practice criteria, or if it moves poor
Friday afternoon projects develop work that can enable a wider group of performers to other projects rather than
Problem solving into major research initiatives. researchers to be brought to bear on the dismissing them, it might limit the range
biggest challenges. These collaborations of its approaches to problem solving and
The best laboratories define a project by Finally, the laboratories we examined are quite different to the transactional undermine performance. And if it does
the specific solution sought, then use a have a healthy attitude to failure, collaborations used to provide a specific, not link its portfolio of work, it also fails to
variety of approaches to solve the problems seeing failed experiments as excellent routine service: they tend to be long- make use of the intellectual scale of the
along the way. They are careful to build learning opportunities. They take term relationships whereby researchers organization and misses opportunities to
on the platforms and techniques in which time to review raw data thoroughly, work in one another’s laboratories and bring new insights to solving problems. It
they have proven strengths in order to see and might invite the head of another meet regularly to share ideas and solve might explain away its decision not to invest
where they can extend into new areas. In laboratory or team to do the same to problems. These collaborations are often in “side projects” by invoking budgetary
this way, their areas of expertise evolve. help tease out any useful conclusions. with groups headed by laboratory alumni, and business pressures, but the outcome
particularly in the academic sphere. remains the same: less enthusiastic
Before running experiments, they ensure Collaboration researchers and missed potential.
they have an initial hypothesis. To avoid Top labs are also aware of how physical Budgetary concerns may also affect
mission creep, they may even write the An open, sharing culture is important for proximity can promote collaboration, the extent to which average labs can
first draft of the final paper abstract at productivity. Lab protocols and manuals organizing departments so that different design their facilities to encourage
the outset—even if they do not intend to are explicitly codified and shared, but teams and disciplines work closely internal collaboration: teams working in
publish the work externally—and redraft top labs also call regular meetings for together and even sit in the same older facilities are often separated from
the paper as work progresses. One teams to share knowledge and challenge areas. In one laboratory, chemists and other parts of their laboratory group,
academic laboratory uses the draft final it, and to discuss the difficulties they biologists share a space for their desk and might even be located on different
paper as the starting point for all reviews. face. They also create opportunities work, with the chemistry lab on one side sites. But again, whatever the cause, the
for teams to meet informally, perhaps and the biology lab on the other. Large result is likely to be lower productivity
Notwithstanding these controls, laboratory over coffee or lunch, seeing this as an coffee areas and a lunch room for the because of lost opportunities to share
heads are keen to give teams a degree important way to identify how different laboratory staff encourage the culture of learning and solve problems together.
of autonomy and not over-define the teams can support one another. sharing and networking between teams.
specific scientific approaches they should One academic laboratory holds a Journal “Hospitals and labs are the opposite: at We are not suggesting that one size fits all.
use, as doing so risks demotivating Abstract Club, at which all abstracts hospitals you want to minimize contact Academic and industry laboratories have
researchers and smothering potentially from the top 30 scientific journals are to prevent diseases spreading; in labs different requirements, and our research
12 Pharma R&D Compendium 2010 13
The secret of high productivity in the research lab

did not identify a single laboratory that top labs take to make their research
followed all the best practices described activities productive, from the bottom up.
here. Nevertheless, the research identifies It should prove a useful resource to those
the common approaches the world’s wishing to emulate their performance.

For more information about the research, analysis, and its application, please contact
successlab@mckinsey.com

Mark Beards (mark_beards@mckinsey.com) is a consultant in McKinsey’s London office,


where Michael Edwards (michael_edwards@mckinsey.com) is an associate principal, and
Mubasher Sheikh (mubasher_a_sheikh@mckinsey.com) is a principal.

We identified 30 internationally prestigious awards in medicine, biology, and chemistry, then


1

identified the recipients of these awards between 2000 and 2008. Their innovation productivity
was measured by their impact-adjusted publication productivity (Hirsh index divided by years
of publishing activity) and IP-generation productivity (US patents divided by years of publication
activity). Twelve of the top-quartile scientists agreed to take part in the research.
Pharma R&D Compendium 2010 15
The future of drug development: advancing clinical trial design

Traditionally, drug development has been conducted in distinct—and


The future of drug development: increasingly expensive—phases. A more integrated model that uses
advancing clinical trial design adaptive design approaches to enhance flexibility and maximize the use
of accumulated knowledge could improve the quality and reduce the cost.

John Orloff, Frank Douglas, Jose Pinheiro, Susan Levinson, Michael Branson,
Pravin Chaturvedi, Ene Ette, Paul Gallo, Gigi Hirsch, Cyrus Mehta, Nitin Patel,
Sameer Sabir, Stacy Springs, Donald Stanski, Matthias Evers, Edd Fleming,
Navjot Singh, Tony Tramontin, and Howard Golub

Pharmaceutical innovation is increasingly relevance for the biological target in a


risky, costly, and at times inefficient—with given disease intervention and insufficient
obvious implications for productivity.1–3 understanding of the dose–response
Estimates of the average cost of bringing relationship of the new molecular entity.
a new drug to market range from $800
million to $2 billion, in part owing to As recognized by the Critical Path
late-stage failures and the rising costs of Initiative of the US Food and Drug
Phase II and III trials.4–9 Conducting these Administration (FDA), novel approaches
phases of development more effectively to clinical trial and program design could
and reducing attrition rates are therefore have an important role in overcoming
major goals. The problem of attrition is these challenges. The traditional
particularly acute in Phase II trials,10 owing approach to drug development separates
to factors such as the lack of proof of clinical development into sequential,

During the exploratory phase of development, this new model uses all available knowledge and tools, including
biomarkers, modeling and simulation, and advanced statistical methodology. Trials are designed to determine
proof of concept (PoC) and to establish dose selection to a level of rigor that will enhance the likelihood of success
in the confirmatory phase.
During the confirmatory
EXHIBIT 1
A novel model for clinical development phase, modern designs, tools,
and knowledge are applied to
larger-scale studies with the
goals of identifying the target
Target PoC Approval
patient population in which the
Target discovery drug is efficacious, establishing
PoC clinical trials Clinical development
and validation
the benefit/risk ratio, and
confirming the optimal dose
Exploratory phase Confirmatory phase
and dosing regimen. During
▪ Apply biomarkers, modeling and simulation, ▪ Apply innovative tools and clinical trial this phase, innovative clinical
and advanced statistical methodology designs such as adaptive or seamless
▪ Demonstrate PoC and establish dose selection studies trial designs such as adaptive
▪ Identify target patient population,
confirm optimal dose and dosing
regimen, and establish the benefit/risk
or seamless studies compress
ratio
timelines, improve dose and
regimen selection, and reduce
the number of patients assigned
to non-viable dosing regimens.
16 Pharma R&D Compendium 2010 17
The future of drug development: advancing clinical trial design

distinct phases, in which progress Exploratory phase 1. Tools, methods, and designs for enhancing development
is measured at discrete milestones, of development
Modeling and simulation
separated by “white space.” We argue
These techniques are a cornerstone of the novel drug-development model. In the exploratory phase,
that the effectiveness of the clinical Modeling is a key feature of the more modeling and simulation can help to refine dose selection and study design, and represent dose-response
development can be improved by integrated approach (Exhibit 1). Biological and time-response behavior of safety and efficacy endpoints. In combination with Bayesian methods,
adopting a more integrated model that modeling is used to understand these can provide a continuous flow of information across different phases of development. Modeling in
increases flexibility and maximizes the use genetic, biochemical, and physiological early development also enables the use of external information (an important issue in the light of current
of accumulated knowledge. In this model, networks, as well as pathways and discussions within the industry about sharing placebo data among companies), which could greatly
broader, more flexible phases leading to processes underlying disease and increase the efficiency of investigations in early development.
submission for approval are designated pharmacotherapy.11,12 Pharmacological
“exploratory” and “confirmatory” (Exhibit modeling guides clinical-trial design, dose In the confirmatory phase, simulation can clarify how different study designs affect the outcome and
1). This model is adaptive, parallel, selection, and development strategies.13,14 likelihood of success, thereby guiding development strategy. In the latter case, this is facilitated by pooling
many sources of data both from prior studies of the drug and external data that might be an informative
and data-led and allows all available Finally, statistical modeling can be used
guide to achieve better decision-making. Furthermore, these techniques can be used not just during the trial-
knowledge to be appropriately shared to assess development strategies and
design process, but also mid-study through the use of adaptive trial designs.
across the breadth of development trial designs in populations.11,12,15 These
studies to improve the quality, timeliness, three types of modeling should be used Bayesian methodology
and efficiency of the process. throughout the development process to This relies on the use of probability models to describe knowledge about parameters of interest (for example,
maximize their impact and synergies. the treatment effect of a drug in development). Bayesian inference uses principles from the scientific method
Central to this model are novel tools, to combine prior beliefs with observed data, producing enhanced, updated information (for reviews, see
including modeling and simulation, In the exploratory phase, modeling and footnotes 22 and 23.) Using Bayesian methodologies, initial beliefs about the parameters are summarized
Bayesian methodologies, and adaptive simulation can help refine dose selection in their prior distribution. Then, new data values are collected experimentally (for example, patient survival
designs such as seamless adaptive and study design. Early development in an oncology trial) and the probability distribution of these values leads to the likelihood function (the
designs and sample-size reestimation studies are conducted with fairly observed evidence on the parameters). The two elements are then combined, using Bayes’ theorem, to
produce the posterior distribution of the parameters—that is, the updated knowledge given the observed
methods (see sidebar 1, “Tools, methods, restricted resources (limited duration,
evidence. By contrast, frequentist methods rely solely on observed evidence for inferences, and typically do
and designs”). These can ensure the sample sizes, and so on), and the use of
not formally take into account prior information.
judicious use of limited patient resources, all available information is thus crucial for
reduce patients’ exposure to ineffective effective decision making.16 However, it Adaptive designs
or poorly tolerated doses, and lead to should be noted that early development In adaptive trial designs, interim data from a trial is used to modify and improve the study design, in a pre-
the recruitment of patients who, on the decisions based on biomarkers that have planned manner and without undermining its validity or integrity. In the exploratory setting, an adaptive
basis of biomarker analysis, are most not been fully qualified can be misguided trial can assign a larger proportion of the enrolled subjects to the treatment arms that are performing well,
likely to respond and represent the most if such biomarkers eventually prove not drop arms that are performing poorly, and investigate a wider range of doses so as more effectively to select
favourable benefit/risk ratio. to correlate with, or be predictive of, the doses that are most likely to succeed in the confirmatory phase. In the confirmatory phase, adaptive design
final outcome. Accordingly, it is important can facilitate the early identification of efficacious treatments, the decision to drop poorly performing trial
Here we describe the general issues and to conduct methodology research in arms, and the decision to terminate the trial for futility, and make sample-size adjustments at interim time
points to ensure that the trial is adequately powered. In some cases, it might even be possible to enrich the
methods involved, and show how the parallel with the development program
patient population by altering the eligibility criteria at an interim time point.
tools can be applied in both exploratory to establish the correlation between the
and confirmatory development by using biomarker and late-stage endpoints or Seamless designs
specific cases in which modern trial outcomes. A seamless design combines, in a single trial, objectives that are traditionally addressed in separate trials.
designs and statistical approaches A seamless adaptive design addresses objectives normally achieved through separate trials using data from
have been successful. We hope to raise Modeling and simulation approaches all trial stages, such as seamless adaptive Phase II/III trials.
awareness of these issues among those can be used to represent dose-response
involved in clinical trials and provide and time-response behavior of safety Sample size reestimation methods
guidelines to ensure that the most and efficacy endpoints. Furthermore, These provide the flexibility to increase or decrease the sample size at an interim point in the trial. This is
appropriate solutions are implemented, these approaches can be combined with important if there is uncertainty about between-subject variance in the response, or about the clinically
with the ultimate goal of increasing the Bayesian methods to provide a continuous meaningful effect size at which to power the trial. These methods allow the study to begin with a certain
sample size that can be increased or decreased at an interim point, and even allow for an efficacy-
efficiency and probability of success in flow of information across different phases
stopping boundary.
clinical development. of development. For example, preclinical
18 Pharma R&D Compendium 2010 19
The future of drug development: advancing clinical trial design

data can be used to construct models Modeling and simulation for dose and dosing regimen that would achieve endpoints and a failure to use all of the
and to provide prior information on model and dose regimen selection. An the desired response for the majority of available longitudinal measurements
parameters. Likewise, the results from important goal of a drug-development patients—in this instance, an 80 percent collected in the study.22
a proof-of-concept (PoC) study can be program is the selection of a dose probability that 90 percent of patients A typical design for efficacy and safety
used to form prior distributions for a similar and dosing regimen that achieves the would remain flare-free for two months. evaluation in a PoC study is to use cohorts
model to be used in a subsequent dose- target clinical benefit while minimizing The data derived from this modeling in a dose-escalation algorithm. Cohorts
finding study.11,12,17,18 undesirable adverse effects. Biological exercise allowed for selection of a dosing are assigned, in sequence, to increasing
and pharmacological modeling can be regimen that was investigated and doses until the maximum tolerated dose
An additional benefit of modeling in early useful in this context.19,20 For example, confirmed in a Phase III trial21 (clinical data (MTD) is reached, or unacceptable safety
development is that it allows the use Novartis has used it in the dose selection on various dosing intervals provided the is observed for a given cohort. A new
of external information (for example, for canakinumab (Ilaris, Novartis), a raw data for the modeling and simulation cohort is allowed to start only when
baseline values for safety endpoints) monoclonal antibody that has recently exercise that finalized the dose and acceptable safety signals are verified for
to estimate characteristics of interest been approved for the treatment of regimen selection for Phase III). Similarly, all previous doses. At the end of the study,
about the population. Given the vast the rare genetic disease Muckle-Wells modeling has been used to predict the the goal is either to determine a dose range
quantity of data from other development syndrome (Exhibit 2). Clinical data on impact of changing the dose or dosing for further exploration in Phase IIb, or to
programs that are available in most the relationship between activity of regimen of a dipeptidyl peptidase IV conclude that no PoC can be established
pharmaceutical companies, as well as the therapeutic target (interleukin 1), inhibitor that is being developed for the based on the efficacy-safety trade-off.
current discussions within the industry markers of inflammation, and remission treatment of type 2 diabetes.
about sharing placebo data across of symptoms were captured in a Because of small cohort sizes, only safety
companies, this has huge potential to mathematical model that was continuously Bayesian modeling combined with use problems occurring in a relatively large
improve the efficiency of investigation in adjusted to fit emerging data. Simulation of external baseline data to improve percentage of patients can be reliably
early development. was then used to propose a suitable dose efficacy and detection of safety signals detected by dose-escalation procedures.
in early development. Early development Likewise, only relatively strong efficacy
studies for establishing PoC often signals can be detected with reasonable
Muckle-Wells syndrome is a rare genetic disorder characterized by fever, urticaria, joint pain, and malaise. A use small patient cohorts (ten to 20 statistical power. The detection of safety
monoclonal antibody against interleukin-1β (IL-1β), canakinumab, has been developed to treat such an IL-1- subjects). These patients are usually and efficacy signals can be made more
dependent inflammatory disease. The antibody is delivered parenterally and binds to free IL-1β, driving it into observed for a relatively short period efficient in various ways: by drawing on
the inactive complex and leading to remission of symptoms.21 Total IL-1β, which represents mainly the inactive (several weeks) to evaluate early efficacy data and information external to the trial,
complex, increases after dosing and can be measured. By the laws of mass action, the free and active form of IL-1β,
and safety signals, which are frequently and deploying longitudinal modeling
which cannot be measured, must decrease. However, the reduction in free IL-1β results in a decrease in markers
measured on a continuous scale. approaches to make use of all available
of inflammation, including
C-reactive protein (which can
However, the endpoints for the decision information. Furthermore, the utility of PoC
to proceed with development or not are studies within drug-development programs
be measured), and a remission EXHIBIT 2
of clinical signs and symptoms Dose selection in the development of a therapeutic typically based on a single time point (for can be enhanced by incorporating the
of disease. The clinical data for Muckle-Wells syndrome example, change from baseline at the information obtained in them directly into
on these relationships can be Dose end of the study) and use dichotomized later-phase trials.11,12 Bayesian modeling
captured in a mathematical Total IL–1β
(complex) versions of the original variables to techniques are particularly useful in
model, shown in the exhibit, characterize responder and non-responder implementing these approaches.
and is continuously adjusted behavior. An example of the latter is the
in the light of new data. This transformation of continuous liver-function Adaptive trial designs in early development.
Antibody
framework simulation could
test measurements (for example, alanine The core concept of adaptive trial design
then be used to propose a Free IL–1β
suppressed aminotransferase [ALT] and aspartate (also known as flexible design) is that it
suitable dose and dosing
regimen that would be
aminotransferase [AST]) into binary uses accumulating data to decide how to
predicted to produce a desired indicators—for instance, exceeding modify aspects of the study mid-trial, in a
response for the majority of C-reactive three times the upper limit of normal pre-planned manner, without undermining
protein
patients (for example, an 80 Flaring (ULN). There are, therefore, two types of the validity or integrity of the study.23–26
percent probability that 90
Remission
Symptoms information loss that often occur in PoC Possible adaptations include adjustments
percent of patients will be flare- -56 0 56 112 168 224 280 336 392 448 504 560 studies: the dichotomization of continuous to sample size, allocation of treatments,
Time (days)
free for two months).
20 Pharma R&D Compendium 2010 21
The future of drug development: advancing clinical trial design

2. Case study: combining proof-of-concept and dose-ranging trials into a single adaptive trial

This example shows how a single adaptive trial can replace two standard trials—PoC and dose-ranging. EXHIBIT A EXHIBIT B
It also shows that the combined trial has greater power than the standard PoC design and is substantially Scenario 1 Scenario 4 Power of Power of
Scenario 2 Scenario 5 standard PoC combined
better at estimating the dose-response curve. Mean response
Scenario 3 Scenario 6 Scenario trial1 adaptive design
25 Scenario 7
1 5 5
The trial evaluated an analgesic drug to treat dental pain and tested seven doses of the drug. Several designs
20 2 33 84
with different sample sizes, randomization ratios of drug to placebo, and starting doses were simulated
3 32 67
against several scenarios. Here, we describe one design with a sample size of 120 subjects (40 placebo, 80 15
4 86 100
drug). Bayesian adaptive trials were simulated over seven drug-response scenarios to enable comparisons 10 5 85 99
with standard designs. Seven scenarios, which represent the gamut of probable dose-response curves, were 6 100 100
5
chosen as shown in Exhibit A. In simulations, it was found that across all seven scenarios, a single adaptive 7 99 100
trial can replace two standard trials (PoC and dose-ranging). The power of the trend test for PoC was 0
1 2 3 4 5 6 7 8 Dose 1 Sample size = 30
always greater for the adaptive design, as shown in Exhibit B.

When there was a small dose-response effect (scenarios two and three), the power of the adaptive design EXHIBIT C EXHIBIT D
Efficiency Standard design (sample size = 120)
was about double that of the standard design. When the effect size was modest (scenarios four and five), Average allocation Adaptive design (sample size = 120)
Efficiency Efficiency Adaptive design (sample size = 60)
the power was increased to almost 100 percent. When effect sizes were large (scenarios six and seven), the
10 10
power was almost 100 percent for both adaptive and standard designs. 20
8 8
15
For the same total sample size, the adaptive combined PoC-dose-finding trial is more efficient than the 6 6
two standard trials in estimating the response at every dose (Exhibit C). The continuous curve shows the 10
4 4
efficiency of the adaptive design relative to the standard dose-ranging design for scenario seven. Efficiency
2 5 2
at each dose is defined as the ratio of the square of the estimation error of the standard design to the square
of the estimation error of the adaptive design. The bars show the number of subjects allocated to each dose 0 0 0
by the adaptive design. These results are computed by averaging the results of 1,000 simulations. 1 2 3 4 5 6 7 Dose 1 2 3 4 5 6 7 Dose

The overall efficiency across all doses is greater by a factor of five, whereas for the sloping part of the dose- The above results are true irrespective of which of the seven scenarios reflects the true dose-response curve. For
response curve (doses four, five, and six), the adaptive design is three times more efficient. In Exhibit D, all seven scenarios for the same sample size, the efficiency of the adaptive design was about five times that of the
the adaptive combined PoC-dose-ranging trial with 60 subjects is as efficient in estimating the response at standard design over all doses. It was three times that of the standard design for estimating dose response in the
every dose as the two standard trials with a combined sample size of 120 subjects. It is also as powerful in sloping part of the dose-response curve. Another way to think about this result is that for half the sample size, the
testing for PoC. adaptive design is as powerful and efficient as the standard approach with two trials.

the addition or deletion of treatment example, a PoC trial can be combined Successful implementation of adaptive trial programing to develop complex algorithms
arms, inclusion and exclusion criteria for with a dose-ranging trial (see sidebar 2, designs requires a number of things. Drug and fast computing platforms.
the study population, adjusting statistical “Case study”). This has the advantage responses need to be rapidly observable
hypotheses (such as non-inferiority or of reducing start-up costs and the time relative to accrual rate; alternatively, good Confirmatory phase of development
superiority), and combining trials or between trials, while potentially increasing longitudinal models can be used to forecast
treatment phases. Adaptive trials have statistical power and improving estimates endpoints in time to adapt dose assignments The primary goals of a confirmatory clinical
the potential to translate into more ethical of dose response. Adaptive designs can for future subjects (assuming, of course, that trial are to ensure that the diagnostic or
treatment of patients within trials, more also enable trialists to work with more the early measurements are good predictors therapeutic intervention causes less harm
efficient drug development, and better use candidate doses without increasing the of the late endpoint values). Adaptive trials than good (safety) and to find efficiently and
of available resources. sample size. This is important to reduce also necessitate more upfront statistical confidently the actual effect size of the chosen
the risk of failure in confirmatory trials, in work to model dose-response curves and to primary outcome(s) within the identified
The standard approach to early which, it is estimated, 45 percent of Phase perform simulations—and many simulations patient population (efficacy). Optimization of
development programs is to separate the III programs industry-wide do not have the are required to find the best combinations of trial design during confirmatory development
trials for PoC, dose ranging, and dose optimum dose.3 Adaptive dose-ranging sample size, the randomization ratio between holds the promise of greater success rates,
selection. Adaptive designs offer several studies are discussed further in sidebar 3, placebo and drug, starting dose, and number improved efficiency, better detection of
benefits over the standard approach. For “Adaptive dose finding.”27, 28 of doses. This in turn demands efficient safety signals, compressed timelines, smaller
22 Pharma R&D Compendium 2010 23
The future of drug development: advancing clinical trial design

overall programs, and lower attrition up time for the endpoint used for selection 3. Adaptive dose finding
rates. A number of novel approaches compared with duration of enrollment.
In an adaptive dose-finding study, the dose assignment to the next subject, or next cohort of patients, is
to confirmatory development that can Shorter follow-up will be more conducive to
based on the responses of previous subjects and chosen to maximize the information about the dose-response
contribute to fulfilling this promise are their use, whereas a relatively long endpoint curve, according to some pre-defined objective metric (for example, variability in parameter estimates).
highlighted below. follow-up period will tend to militate against. In a traditional dose-finding trial, selecting a few doses might not adequately represent the dose-response
Development programs that do not involve relationship and many
Seamless adaptive designs. Efficiency complex treatment regimens might thus ADAPTIVE DOSE FINDING patients will be allocated
can be increased through the use of lend themselves better. Drug supply and to “non-informative” doses
Response
seamless adaptive designs, which aim to drug packaging will be expected to be more (wasted doses), as shown
combine objectives traditionally addressed challenging in this setting. in the exhibit. In adaptive
in separate trials in a single trial.25,29 An dose-finding, the strategy
example is the seamless adaptive Phase II/ A number of logistical and regulatory actions is initially to include only
a few patients on many
III design addressing objectives normally must be taken to avoid compromising an
doses to explore the dose
achieved through separate Phase II and III adaptive trial. First, the actual algorithm for
response, then to allocate
trials. These trials are confirmatory in nature, deciding the adaptation to implement must the dose range of interest
as opposed to seamless adaptive trials in be specified in advance. This is usually “Wasted” doses Dose “Wasted” doses
to more patients. This
early development, which are essentially accomplished by creating a charter for the reduces the allocation
exploratory. The first stage of a seamless independent data-monitoring committee of patients to non-informative doses.27,28 Compared with fixed randomization, this approach has the ethical
adaptive Phase II/III trial might be similar charged with performing the unblinded advantage that fewer subjects are assigned doses that are too high or too low. It can also avoid additional,
to a late-Phase II trial, with a control group interim analysis and communicating as separate trials that might be necessary when fixed dose-finding trials do not adequately define the dose range.
and several treatment groups (for example, appropriate with the sponsor. In addition,
different dose levels of the same treatment). the sponsor must have developed in-house Adaptive dose-finding trials also require an infrastructure that enables the rapid communication of
Results are examined at the end of the first procedures to ensure that the algorithm is responses from trial sites to a central, unblinded analysis center and of adaptive dose assignments to the
trial sites. Randomization software capable of rapidly computing dynamic allocation of doses to subjects
stage, and one or more of the treatment not transmitted throughout the company,
is additionally mandated by adaptive trials because pre-specified randomization lists will not work. In
groups are selected to continue, along with and especially not to the study investigators.
addition, a flexible drug-supply process is required because demand for doses is not fixed in advance, but
the control group, into the trial’s second rather evolves as information on responses at various doses is gathered as the trial progresses.
stage. The final analysis comparing the To maintain a trial’s integrity, the processes
selected group(s) with the control will use by which interim data are examined
data from the continuing groups from both and selection decisions are made and
stages of the trial. implemented must be considered carefully. final analysis uses only data from patients Other considerations for adaptive designs
Current conventions that restrict knowledge enrolled after the selection decision. This include the endpoint used for selection.
There are three main potential advantages of interim results in ongoing trials should might allow a broader investigation of the This need not be the same as the endpoint
of seamless adaptive designs: they shorten be respected to avoid compromising the first-stage data involving the sponsor’s to be used in the main study analysis;
the clinical-development program by “interpretability” of trial results. In some personnel, while alleviating concerns if a good surrogate marker is available,
eliminating the time lag between Phase II cases the decision being made at the about the trial’s integrity; in addition, this can be used and might enhance the
and III trials; they lead to greater efficiency selection point of a seamless design will traditional non-adaptive statistical efficiency of the seamless trial. Second,
in the use of data from both stages, which be one for which a sponsor’s perspective methodology normally suffices. Such modeling and simulation will probably
might mean that fewer patients are required might be relevant and for which the designs may maintain the advantage have an important role in developing
to obtain the same quality of information; sponsor traditionally has been responsible, of reducing “white space,” while losing the specific details of seamless designs
and they enable the earlier acquisition of raising the question of its involvement the efficiency that results from using (for example, per-group sample sizes in
long-term safety data, gathered through in the monitoring process. A distinction data accrued in stages. Regardless, the different stages, considered under
continued follow-up of patients from the is sometimes made between seamless operating procedures for the monitoring various scenarios) to ensure that they
first stage.25,29 adaptive designs that are inferentially process in seamless designs must be are robust and efficient. Third, the final
seamless and those that are operationally carefully considered to ensure that the analysis must use statistical methodology
The designs are not suitable for all seamless. In the former, which we describe right expertise is applied to the decision, that is appropriate for the design: “naïve”
drug-development programs. Feasibility here, the main analysis uses data from while limiting access to the accruing data comparisons of control versus the selected
considerations include the length of follow- both stages of the trial. In the latter, the as appropriate to maintain integrity. treatment that do not account for the
24 Pharma R&D Compendium 2010 25
The future of drug development: advancing clinical trial design

4. Issues with the standard clinical-development approach methodology has several drawbacks. If confirmatory trials, still requires significant
the actual effect is substantially larger than resources to perform appropriately. Also,
Issues with the standard approach to clinical development can be illustrated by considering a randomized
the SCID, a smaller sample size would the inevitable start-up time and wind-
clinical trial with the following assumptions. Based on available evidence from early-phase trials, it is
estimated that σ = 1, that the anticipated effect size δ = 0.2, and that the smallest clinically important delta
have sufficed to attain adequate power.32 down activities between trials have to be
(SCID) is 0.1. Rather than conservatively enrolling a sample size required to demonstrate the SCID (4,000 Sponsors will not often risk significant included when deciding true program
subjects), the sponsor appropriately powers the trial to detect the estimated larger δ (1,000 subjects). Now, resources on trial sizes based on SCID efficiency and development timelines. This
suppose that the true underlying value of δ is 0.15. In that case, a sample size of 2,000 subjects would be assumptions that would lead to larger might therefore not be the most efficient
required to power the trial adequately to detect this difference. The difficulty is, of course, that the true trials than the current “best guess” about way to proceed from the viewpoint
underlying value of δ is not known at the start of the trial. In this example, the 1,000-subject study would the actual effect size (see sidebar 4, of the entire clinical-trial program.
probably yield a non-significant result, as it is powered only to detect an effect size of 0.2, which is larger “Issues with the standard approach”).
than the actual effect size of 0.15. Instead, a smaller trial corresponding Advantages of adaptive SSR in
to that best guess may be run; if that confirmatory trials. A more flexible
In this example, unless the 1,000-patient, under-powered trial were repeated with a larger sample size, a
assumption is too optimistic, and the truth approach to the fixed sample-size
potentially efficacious treatment would be unnecessarily and unfortunately discarded. If the trial were to be
is an effect size closer to the SCID, the methodology is needed. By altering the
repeated with the reestimation of the actual effect size, then 2,000 patients would need to be enrolled, and
the time and resources to perform the original trial (sometimes more than three years) would have been
trial will be underpowered and thus have a sample size using interim data from the
spent without much benefit other than to gain a more reliable estimate of the actual effect size in order to high chance of failure. trial itself, this flexibility can be achieved
design the second trial. More importantly, the subjects for that study would have been put at unnecessary without compromising the power or the
risk because the study had no real chance of being definitive. One approach to solving the problem false-positive rate of the trial (that is, the
of uncertainty about δ is to design chance of making a false claim of efficacy
and execute further exploratory trials for a treatment that is not efficacious). SSR
design will not be appropriate. Finally, the treatment and control arms with respect (typically Phase II studies). These small should be considered in two situations:
aptness of the design does not depend to the primary endpoint. Even though Phase II studies are normally carried where there is significant uncertainty about
on any particular algorithm for choosing the true value of δ is unknown, the trial out to get a more precise estimate (or σ; or where there is a substantial difference
the patient group to be continued; it is not investigators will usually have in mind a best guess) of the actual δ and σ so between the sample size resulting from
even necessary for a firm algorithm to be specific value, δmin, which represents the that the confirmatory study might be using the SCID and the sample size the
specified in advance, although the general smallest clinically important delta (SCID) adequately powered. Each exploratory sponsor can justify on the basis of its best
principles that will govern the decision for this trial. Next, the trial designers will trial, although somewhat smaller than guess of the effect size.29
should be clear in advance. decide the sample size that can detect
values of δ, based on prior information,
Sample size reestimation within a that exceed the SCID with good power. This is a hypothetical example of a study in which sample size reestimation owing to uncertainty about σ led to
confirmatory trial (Phase III). Sample The standard deviation σ (between an increase in sample size to
size reestimation (SSR) provides a subject variability) is a “nuisance ensure 90 percent power was
EXHIBIT 3 maintained. At the beginning
mechanism for the appropriate use parameter” the true value of which must Reestimating sample size while maintaining statistical power
of the information obtained during be estimated in order to proceed with the of the trial, the planned sample
size was estimated at 150
a confirmatory study to inform and sample size calculation.
patients based on a standard
adjust the necessary sample size going Δ = 0.375
Power – 90%
Δ = 0.375
Power – 90%
deviation of 1.0. At the interim
forward.30,31 This process increases the The SCID can be often pre-specified σ = 1.0 σ = 1.4

N = 150 N = 295 LPFV 1 analysis, the actual standard


confidence that an appropriate sample from purely clinical arguments, whereas deviation was 1.4. Even though
size has been chosen to answer the the actual effect size is unknown. Thus it Learning
the effect size was as originally
primary study questions. is possible in principle to design a study Active predicted, an increase in
with a fixed sample size that will have sample size to 295 patients
The standard approach used to power adequate power to detect the SCID, in Control
would be required to maintain
a confirmatory study is first to estimate the absence of adequate prior information 90 percent power. Without
the underlying treatment effect of the about the actual effect size of the test Enrollment the sample size reestimation,
Interim analysis Final analysis
primary endpoint based on available prior agent. This is what statisticians envisaged Sample size the power at the final analysis
reestimation
would be only 64 percent and
information. The parameter δ denotes the when they created the fixed-sample
there would be greater risk of a
true underlying difference between the methodology. However, this fixed-sample
1Last patient first visit. failed trial.
26 Pharma R&D Compendium 2010 27
The future of drug development: advancing clinical trial design

5. Group-sequential and adaptive designs for sample size reestimation

Group-sequential design about efficacy and safety and then following it up with a larger Phase III trial once the efficacy and safety of the
Suppose that the sponsor is unsure of the true value of δ, but nevertheless believes that it is larger than the smallest compound have been established. There is, however, an important distinction between the conventional Phase II
clinically important delta (SCID). In this case, a group-sequential design might be considered. Such a design is followed by Phase III strategy and the adaptive strategy outlined below.
characterized by a maximum sample size, an interim monitoring strategy, and a corresponding boundary for
early stopping for efficacy. The maximum sample size is computed so that the study has adequate power to detect a In the conventional approach, the data from the Phase II trial are not combined with the data from the Phase
value of δ that the sponsor believes represents a reasonable estimate of the efficacy of the experimental compound, III trial. The adaptive design, however, uses all the data from both stages for the final analysis. This can have
provided this estimate is at least as large as the SCID. If the sponsor wishes to be very conservative about this important advantages both in terms of gaining additional statistical power and in shortening the drug-
estimate, the maximum sample size needed can be computed to have adequate power at the SCID itself. An upfront development time.
commitment is made to enrol patients up to this maximum sample size. However, if the true δ exceeds the SCID, the
trial might terminate earlier with high probability by crossing an early stopping boundary at an interim analysis. In our example, we stated that the SCID was 0.1. Supposing that the sponsor believes that the true δ = 0.2—that is,
twice the size of the SCID. If this is indeed the case, then a total sample size of 1,000 patients will have 89 percent
Returning to the example discussed in Sidebar 4, suppose that the sponsor decides to make an upfront commitment power at a one-sided α level of 0.025. On this basis, the sponsor is prepared to make an initial investment of 1,000
of 4,000 patients to the trial but intends to monitor the accruing data up to four times, after 1,000, 2,000, 3,000, patients to this trial. As an insurance policy, however, the sponsor intends to take an interim look at the accruing
and 4,000 patients become evaluable for the primary endpoint. The commitment of 4,000 patients ensures that the data at the mid-point of the
trial will have 88 percent power to detect a difference as small as δ = 0.1 (in this case the SCID). Although this is a Interim estimate, Conditional power without sample size Total sample size needed to achieve trial, after 500 patients are
δ increase, % 80% conditional power
rather large sample size to commit to the trial, the actual sample size is expected to be substantially smaller if the evaluable for response. If the
true δ is larger than the SCID. This is because at each of the four interim monitoring time points there is a chance 0.2 95 720 (sample size reduction) estimate of δ obtained from
of early termination and a declaration of statistical significance. At each interim analysis, a test for statistical 0.175 86 890 (reduction)
these 500 is smaller than
significance using all available primary endpoint data would be performed, and the result would be compared with the sponsor expected, then
0.15 72 1166 (increase)
a properly determined early-stopping boundary value. The trial could be terminated the first time a boundary is the sponsor might choose to
reached, with a valid claim that the experimental arm is more efficacious than the control arm. 0.125 51 1757 (increase) increase the sample size to
preserve the power of the trial.
0.1 30 2990 (increase)
However, sometimes a sponsor might not be willing to make such a large upfront commitment, particularly when
the only currently available data on δ come from one or two small Phase II trials. The sponsor might feel more Many different criteria can
comfortable with a design that starts out with a smaller sample size of, say, 1,000 patients, with the opportunity to be used to decide whether an increase in sample size is warranted. A commonly used criterion is “conditional
increase the sample size at an interim time point and after observing data from the trial itself. This is the motivation power.” The conditional power at an interim analysis is the probability, given the observed data, that the
for the adaptive design considered next. experimental compound will demonstrate efficacy on completion of the trial. The conditional power computation
requires specifying a value for δ. Either the value specified at the initial design stage or the value estimated
The adaptive design from the interim data can be chosen. In this example, we use the interim estimated value of δ for evaluating
The group-sequential design described above is characterized by pre-specifying a maximum sample size upfront conditional power. The table below displays conditional power for various estimated values of δ at the interim
and terminating earlier if the true δ is larger than anticipated. By contrast, an adaptive design pre-specifies a time point, along with the total sample size needed to achieve 80 percent conditional power at the final analysis.
smaller initial sample size, but with the possibility of increasing the commitment after seeing interim data from The entries in the table assume that σ = 1. Note that the final sample size required to achieve 80 percent
the trial. On the surface, this is similar to the usual practice of first running a small Phase II trial to obtain an idea conditional power could increase or decrease from the initially planned sample size of 1,000.

SSR usually involves the choice of a suitable if the accruing data suggest that the large assumption that the standard deviation σ sample size in the situation of unknown
initial sample size, including one or more sample is not needed. The adaptive SSR is known, they apply equally for the case σ: blinded and unblinded. In the instance
interim analyses at which the sample size strategy proceeds in the opposite direction, of unknown σ.30–32 One can start out with of blinded sample size reestimation, the
will be reassessed.30 There are two distinct starting out with commitment to a smaller an initial estimate of σ and a corresponding sponsor uses pooled data to estimate
strategies—the group sequential strategy initial sample size but with the option to sample-size estimate. Then, following an σ. This is permitted with no penalty to
and the adaptive SSR strategy—for increase it should the accruing data suggest interim analysis, one can reestimate this the analysis criteria (that is, alpha, or the
choosing the initial sample size, and then that an increase is warranted30–33 (see sidebar nuisance parameter, enter the updated probability of Type I—false positive—
altering it on the basis of data obtained at 5, “Group-sequential and adaptive designs”). estimate into the equation, and recompute error). It is preferable that the sponsor
various interim analysis time points. The the sample size. An example is given in pre-specifies how many times changes are
group sequential strategy, which is also an Extending the methodology to unknown σ. Exhibit 3. to be made to the sample size, at what
adaptive design, begins with commitment Although the group sequential and adaptive time points, and how the new sample
to a large upfront sample size and cuts back SSR methods were presented under the There are two ways to obtain the new size will be calculated. Usually, this type
28 Pharma R&D Compendium 2010 29
The future of drug development: advancing clinical trial design

of adjustment will not be permitted by considerations to verify their operating data standards. These problems solutions. To address this challenge, we
regulatory authorities more than once. characteristics, and therefore tend to are compounded by competitive make the following recommendations.
For unblinded sample size reestimation, the require more time for the planning and hurdles to sharing what is considered First, a common vocabulary and a
sponsor sets up a mechanism (possibly protocol-development phase. Regulatory proprietary information about novel common understanding of the value of
with the data-monitoring committee agencies and institutional review boards therapies without PoC, which inhibits modern trial designs to all stakeholders
of the trial) whereby the SSR is based also need to approve the design format for the exchange of data. Overcoming must be defined and disseminated.
on an unblinded estimate of variability interim analysis, and these discussions can internal resistance and aversion to Second, guidelines and case studies
(or statistical information) at the interim take time too. Such time considerations change also represents a major hurdle for assessing situations in which tools
analysis. Sample size may be altered one can lead a company to follow the for incorporating the prospective use of should—or should not—be applied must
or more times, but the maximum statistical traditional route to clinical development, novel trial designs and methodologies, be developed and disseminated. Third,
information must be pre-specified. without fully appreciating the advantages and of modeling and simulation, into there is a need for a methodology for
that adaptive designs can eventually bring clinical-development programs. dialogue with regulatory authorities to
If the sponsor agrees that there will be in terms of time and cost savings and facilitate discussion of clinical strategies
no early stopping for efficacy following probability of success. A key barrier to the implementation of which use these tools and address
an interim analysis, then no adjustment tools and techniques which advance the potential constraints. Finally, it will be
to the final analysis criteria is necessary. As described above, adaptive designs quality, timeliness, and efficiency of drug crucial to identify specific solutions to
The data-monitoring committee might further require the following: quickly development is the ability to work across address obstacles and objections that
monitor the data one or more times and observable responses relative to the disciplines and among stakeholders to inhibit the adoption of modern tools and
adjust the sample size up or down based patient accrual rate, or good longitudinal understand how and when to apply these adaptive study designs.
on the unblinded estimate of variability forecasting models; efficient design
and attempt to reach the pre-specified and implementation software and fast This article, with supplementary information, was originally published by Nature Reviews Drug
maximum information. computing platforms; an infrastructure Discovery, and can be found for citation purposes online at www.nature.com/reviews/drugdisc
that facilitates rapid communication, both (doi:10.1038/nrd3025). Please direct scientific correspondence to John Orloff (john.orloff@novartis.
When the sponsor pre-specifies the interim across trial sites to the central unblinded com). Requests of a general nature can also be made to the McKinsey Trial Design Team (matthias_
time points at which it is permissible to analysis centre, and of dose assignments evers@mckinsey.com).
terminate early for efficacy, the criteria for to trial sites; and a flexible drug-supply
each interim analysis must be pre-specified process. Appropriate models, which John Orloff, Jose Pinheiro, Michael Branson, Paul Gallo, and Donald Stanski are at Novartis
in a manner that controls the false-positive reliably characterize the longitudinal Pharmaceuticals Corporation; Frank Douglas is at the Kauffman Foundation; Susan Levinson is
rate across the entire study. This will result behavior of clinical endpoints, or the at The Strategic Choice; Pravin Chaturvedi is at Napo Pharmaceuticals; Ene Ette is at Anoixis
in adjustment to the final analysis criterion relationship between biomarkers and Corporation; Gigi Hirsch, Nitin Patel, Sameer Sabir, Stacy Springs, and Howard Golub are at
if the study is not stopped early. Interim endpoints, are also crucial to the success the Center for Biomedical Innovation, Massachusetts Institute of Technology; Cyrus Mehta is at
looks undertaken solely for administrative of the modern clinical-development Cytel Statistical Software and Services; and Matthias Evers (matthias_evers@mckinsey.com),
purposes, with no intention of stopping paradigm discussed here. Because Edd Fleming (edd_fleming@mckinsey.com), Navjot Singh (navjot_singh@mckinsey.com), and
the trial in the light of efficacy data, do not model assumptions often need to be Tony Tramontin (tony_tramontin@mckinsey.com) are at McKinsey & Company.
need to have defined criteria. The trial then checked—and at times revised—after
proceeds either until it is terminated early data have been observed, an intriguing We would like to acknowledge W. Dere (Amgen), S. Cummings (UCSF), A. Lee (Pfizer), and E.
for efficacy on the basis of the pre-defined possibility would be to use “adaptive Berndt (MIT-CBI) for significant contributions to discussions leading to this manuscript.
criteria having been reached, or until the modeling” approaches. This is a topic for
planned maximum information (sample size further research.
or number of events) is reached.
Maximizing the use of all potential prior
Tackling the challenges information requires greater collaboration
of adaptive trial designs across functional silos in organizations
to avoid compartmentalization of
Because they are so flexible, these new data. In practice, the inclusion of a
trial designs require significant statistical broader sample of data sets can be
analyses, simulations, and logistical difficult because of the lack of common
30 Pharma R&D Compendium 2010 31
The future of drug development: advancing clinical trial design

1
Booth, B. and Zemmel, R., “Prospects for productivity,” Nature Reviews Drug Discovery, 3, 25
Krams, M. et al., “Adaptive designs in clinical drug development: opportunities, challenges, and
451–456 (2004). scope reflections following PhRMA’s November 2006 workshop,” J. Biopharm. Stat. 17, 957–964
2
Gilbert, J., Henske, P., and Singh, A., “Rebuilding big pharma’s business model,” In Vivo 21, 1−4 (2007).
(2003). 26
Gallo, P. et al., “Adaptive designs in clinical drug development—an executive summary of the
3
Kola, I. and Landis, J., “Can the pharmaceutical industry reduce attrition rates?” Nature Reviews PhRMA Working Group,” J. Biopharm. Stat. 16, 275–283 (2006).
Drug Discovery, 3, 711–716 (2004). 27
Bornkamp, B. et al., “Innovative approaches for designing and analyzing adaptive dose-ranging
4
Budget US Government, App., FY 1993–2003. trials,” J. Biopharm. Stat. 17, 965–995 (2007).
5
Parexel’s Pharmaceutical R&D Statistical Sourcebook 2002/2003 (Parexel International, Waltham, 28
Weir, C. J., Spiegelhalter, D. J., and Grieve, A. P., “Flexible design and efficient implementation of
US, 2003). adaptive dose-finding studies,” J. Biopharm. Stat. 17, 1033–1050 (2007).
6
Adams, C. P. and Brantner, V. V., “Spending on new drug development,” Health Economics, 29
Bretz, F., Schmidli, H., König, F., Racine, A., and Maurer, W., “Confirmatory seamless Phase II/III
February 26, 2009 (doi: 10.1002/hec.1454). clinical trials with hypotheses selection at interim: general concepts,” Biom. J. 48, 623–634 (2006).
7
DiMasi, J. A., Hansen, R. W., and Grabowski, H. G., “The price of innovation: new estimates of 30
Mehta, C.R. and Patel, N.R., “Adaptive, group sequential and decision theoretic approaches to
drug development costs,” J. Health Econ. 22, 151−185 (2003). sample size determination.” Stat. Med. 25, 3250–3269 (2006).
8
Adams, C. and Brantner, V. V., “Estimating the cost of new drug development: is it really $802 31
Chuang-Stein, C., Anderson, K., Gallo, P., and Collins, S., “Sample size reestimation: a review and
million?” Health Aff., 2, 420–428 (2006). recommendations,” Drug Inf. J. 40, 475–484 (2006).
9
Pharmaceutical R&D Factbook 2007 (CMR International, London, UK, 2007). 32
Golub, H. L., “The need for more efficient clinical trials,” Stat. Med. 25, 3231–3235 (2006).
10
KMR General Metrics Study (KMR Group, Chicago, US, 2007). 33
Tsiatis, A. A. and Mehta, C., “On the inefficiency of the adaptive design for monitoring clinical
11
Sheiner, L. B. and Steimer, J. L., “Pharmacokinetic/pharmacodynamic modeling in drug trials,” Biometrika 90, 367–378 (2003).
development,” Ann. Rev. Pharmacol. Toxicol. 40, 67–95 (2000).
12
Lalonde, R. L. et al., “Model-based drug development,” Clin. Pharmacol. Ther. 82, 21–32 (2007).
13
Breimer, D. D. and Danhof, M., “Relevance of the application of pharmacokinetic-
pharmacodynamic modeling concepts in drug development. The ‘wooden shoe’ paradigm,” Clin.
Pharmacokinet. 32, 259–267 (1997).
14
Danhof, M., Alvan, G., Dahl, S. G., Kuhlmann, J., and Paintaud, G., “Mechanism-based
pharmacokinetic/pharmacodynamic modeling—a new classification of biomarkers,” Pharm. Res.
22, 1432–1437 (2005).
15
Holford, N. H. G., Kimko, H. C., Monteleone, J. P. R., & Peck, C. C., “Simulation of clinical trials,”
Ann. Rev. Pharmacol. Toxicol 40, 209–234 (2000).
16
Miller, R. et al., “How modeling and simulation have enhanced decision making in new drug
development,” J. Pharmacokinet. Pharmacodyn. 32, 185–197 (2005).
17
Berry, D. A., “Bayesian statistics,” Med. Decis. Making 26, 429–430 (2006).
18
Müller, P., Berry, D. A., Grieve, A. P., Smith, M. and Krams, M., “Simulation-based sequential
Bayesian design,” J. Stat. Plan. Inference 137, 3140–3150 (2007).
19
Goggin, T. et al., “Modeling and simulation of clinical trials: an industrial perspective,” Simulation
for Designing of Clinical Trials (eds. Kimko, H. C. and Duffull, S. B.), 227–224 (Marcel Dekker, New
York, US, 2002).
20
Reigner, B. G. et al., “An evaluation of the integration of pharmacokinetic and pharmacodynamic
principles in clinical drug development. Experience within Hoffman La Roche,” Clin.
Pharmacokinet. 33, 142–152 (1997).
21
Lachmann, H. J. et al., “Use of canakinumab in the cryopyrin-associated periodic syndrome,” N.
Engl. J. Med. 360, 2416–2425 (2009).
22
Berry, D. A., “Bayesian clinical trials,” Nature Reviews Drug Discovery 5, 27–36 (2006).
23
Berry, D. A., “Introduction to Bayesian methods III: use and interpretation of Bayesian tools in
design and analysis,” Clin. Trials 2, 295–300 (2005).
24
Berry, D. A., “Adaptive trial design,” Clin. Adv. in Hematol. Oncol. 5, 522–524 (2007).
Pharma R&D Compendium 2010 33
Using IT to speed up clinical trials

The first wave of IT improvements solved some


Using IT to speed up clinical trials problems, but others remain. Now it is time for a
more comprehensive approach.

Sam Marwaha, Samir Patil, and Navjot Singh

Clinical trials are costly and complex their designs for trials from one stage
undertakings for pharmaceutical to the next. They have improved their
companies. A trial can cost hundreds technological capabilities—especially
of millions of dollars and require the connectivity—at clinical locations,
coordination of many patients, physicians, enabling trial managers to keep tabs on
and regulatory agencies for up to seven the retention of patients and the progress
years. The stakes are high, because of trials. And they have enhanced their
companies spend about 10 percent of ability to conduct trials worldwide by
their operating budgets on these trials vastly increasing the pool of researchers
and, more importantly, can derive huge and patients. They have also adopted
benefits from being first to market with a more disciplined procedures for
new type of drug. But they face intense managing trials, in some cases borrowing
competition from similar compounds and techniques (including “stage gates,”
from generic drugs once their patents run which set firm deadlines for gathering
out. As patents are issued before a drug data and refining the goals of subsequent
goes into clinical trials, the faster a trial stages) from product design.
goes, the longer the manufacturer enjoys
a monopoly until generic versions can Overall improvements in the performance
be sold. Thus, a streamlined and speedy of trials remain elusive, however,
trials process can have a significant especially in the early stages. There are
impact on the economics both of new several reasons. First, many companies
drugs and of existing drugs that may be fail to coordinate trials across their
approved for additional uses after further organizations as well as they could,
clinical studies. and the lack of cross-trial transparency
can create delays when different trials
Over the past decade, pharmaceutical compete for scarce resources. Second,
companies have introduced a number many organizations have yet to embrace
of initiatives to boost the productivity reusability by streamlining their approach
of trials. They have rolled out electronic to the design of trials; for example, certain
data capture (EDC) systems, allowing components of the forms that guide
patients and researchers to enter trial researchers in trials could be shared and
information directly in electronic diaries or reused in other trials. Third, although
online systems. They have adopted new EDC systems have substantially reduced
scientific approaches, such as Bayesian the time required to gather data—to two
techniques1 that enable them to refine weeks, in some cases, from 20—the first
34 Pharma R&D Compendium 2010 35
Using IT to speed up clinical trials

generation of systems EXHIBIT 1 Developing an integrated plan The transformation from ad hoc
was not always flexible Where IT can help planning to an integrated approach
A “clean-sheet” approach can integrate IT systems in a redesign of the whole clinical-trials program
or reliable enough to suit Pharmaceutical companies design trials requires a comprehensive program
investigators’ needs. Recruiting
patients and
within individual therapeutic areas, but that addresses the cultural, process,
Trial phases Design, planning Start-up Close-out Report
investigators,
Fourth, verifying electronic managing trial trials in all of them draw from a pool of and systems barriers to change. Five
data to ensure that they Key activities ▪ Designing ▪ Distributing drugs ▪ Monitoring ▪ Entering, verifying shared functions. Coordinating these elements are vital.
benefiting protocols and information progress, adverse data
correlate with physical from
management
▪ Creating
regulatory
▪ Setting up data
collection
events
▪ Monitoring,
▪ Processing
clinical-response
resources is not easy given the dynamic
data (such as laboratory system documents
▪ Planning, ordering
▪ Selecting site managing drug
supply
forms
▪ Addressing
nature of the trials portfolio, the challenge 1. Support from senior managers, who
drug supplies ▪ Tracking patient investigators’
test reports) remains time enrollment queries of creating the right cross-functional teams must insist, very visibly, on the use of
▪ Tracking clinical-
consuming. Finally, the response forms from the shared pool, and the difficulties standardized processes and systems.
IT focus
productivity measures of recruiting investigators and patients. To
1. Clinical-data ▪ Database of ▪ Standard interface ▪ Automated data
some companies have management investigators and to integrate third- checks to complicate matters, program managers for 2. A common process, that all project
their preferences party pay systems minimize queries
implemented apply only to to aid the design
of electronic forms
quickly and
inexpensively
trials often try to reserve more resources managers follow, for formalizing the
parts of the organization 2. Safety-data Real-time monitoring and analysis of study data to spot adverse reactions to a drug than they need in order to ensure that way the company creates, approves,
management
and thus fail to capture all 3. Document Single repository of data with version control, work flow management
the actual requirements will be available. controls, executes, and ends trials.
of the potential benefits. management
What is more, when trials are canceled,
4. Clinical-trials ▪ Modular design, ▪ System to convert ▪ Electronic invoicing ▪ Report builder
management construction of
consent and case
study designs to
electronic forms
▪ Automated drug
supply work flow
newly freed-up resources often are not 3. A flexible approach to planning
In recent years, some report forms and databases
with minimal
▪ Study planning and
budgeting
redistributed efficiently to ongoing efforts. that enables project teams to
reworking and ▪ Patient
leading companies manual effort management These factors can make the use of a plan at whatever level is clear to
have begun to ease the 5. Project and ▪ Standard data Ability to track and analyze costs, quality, and speed company’s resources less than optimal, them—an entire program, a project
resource models to ease
bottlenecks and raise management coordination with limiting its ability to initiate new studies within a larger program, or a
third-party
productivity by revisiting contractors or to complete ongoing ones quickly. bundle of tasks within a project.
IT systems to streamline Developing an integrated plan across
the process further. therapeutic areas to address these 4. Forecasts of resource use based
Such efforts are most successful when information and process design that are issues can cut costs and speed up the directly on project plans.
companies take a comprehensive, ripe for improvements to make clinical completion of trials: one large company
“clean-sheet” approach, integrating their trials faster and cheaper. identified potential savings of $50 5. Common standards for detailed
systems in a redesign of the whole trials million to $150 million from improving descriptions of work activities that will
program (Exhibit 1). The application of Planning. Integrated planning of clinical the way it allocates resources across help resources to be allocated more
these principles has speeded up some trials improves the way resources (for projects (Exhibit 2). efficiently and specifically.
trials by 10 percent or more. In addition, example, clinicians and biostatisticians)
a number of companies have applied are allocated to clinical teams. EXHIBIT 2 Each of these elements
principles from lean manufacturing Where the savings are has implications for
The business case for addressing planning barriers can be compelling
to these information flows, improving Tools. Modular designs and reusable information technology. IT
their throughput and reducing waste components enable the rapid creation of platforms for enterprise
Estimated savings, %
by redesigning their clinical trials electronic case-report forms. project management2 help
Single, common
▪ Reduction of unnecessary labor,
processes, staff responsibilities, and plan of record
external expenses, and
wasted supplies
20–30 a company to develop and
technology. Certain companies have Use of tools. Physicians can be helped manage a more integrated
enhanced their speed, quality, and to use EDC tools more effectively Standard
processes


Increased efficiency of labor
Better capacity management
10–20 approach. In addition to
costs by focusing on getting the through the provision of training linking available resources
right data the first time, managing and customer support, and the Cross-project


Reduced cycle times
More transparency across portfolio 10–50
and trial schedules
coordination

work flows to reduce bottlenecks, introduction of a standard interface. Increased accountability
automatically, the platform
and making data more transparent Improved ▪ More efficient planning
serves as a repository
20–40

across the entire clinical process. Integrated architecture for information forecasting Better capacity management
of design templates
systems. This will make trials visible to for trials, enforces
Through our work with these companies, management from start to finish, and thus 100% = $50 million to $150 million 100
standards, and helps to
we have identified four areas in more efficient, by eliminating bottlenecks. improve work flows.
36 Pharma R&D Compendium 2010 37
Using IT to speed up clinical trials

Designing trials rapidly information was collected manually, so it The pharmaceutical industry could improve should become more active problem solvers
through modularization was easy for the person recording it to its ability to sign up physicians by making who manage the relationship between
spot and correct obvious errors early in data-collection systems more friendly to the organization conducting the trial and
One result of integrated planning across the process. Programing EDC systems to them in several ways. First, companies the investigators. As part of this shift, their
therapeutic areas is that companies are identify these errors, through a series of should gather and track physicians’ incentives should reflect not just the number
beginning to standardize their processes checks, is time consuming and generates preferences in order to understand how they of visits they make to sites but also their
and tools for designing and recording additional start-up costs for trials. want to use electronic forms. They should speed in recruiting patients and the quality of
a trial, and for guiding the investigators Standardized error-checking modules, by then ensure that the systems are flexible the data gathered.
who conduct it. Designing a trial begins contrast, allow such checks to be built enough to accommodate these preferences.
with setting its aims (that is, what the once and reused many times. If some leading researchers insist on staying Streamlined work flow
researchers hope to prove about a with pen and paper, designers of trials must and maximum trial visibility
new drug) and describing the type of Third, collecting data in a standard form make sure that there is a solid process to
patients and physicians it will require. The makes it easier to perform analyses early convert the data to standard electronic forms The fragmented IT systems and data
designers then work with colleagues— in a trial. This in turn enables project (perhaps in a low-cost remote location). architecture3 of most pharmaceutical
within the company and in partner managers to see initial findings more companies make it extremely difficult
organizations such as contract research quickly and to make interim decisions Likewise, companies should standardize for managers to track productivity
bodies that help coordinate trials—to about the rest of the trial. the way they collect data not only within throughout a trial. Managers might
convert these aims into a series of their organizations but also across be able to reduce bottlenecks in the
documents, including the patient consent Tailoring electronic the industry (see sidebar, “A Sabre for availability of patients, for example, if
form, the list of laboratory tests to be data collection to pharmaceuticals?”). This move will shorten they could see the patient-recruitment
conducted, and the case report form that investigators’ needs the learning curve for physicians as they “pipeline.” But although they have access
physicians fill out during the trial. will have to master the electronic-entry to the clinical-trials management system,
Once the design is completed, the system once only. information on patient recruitment is
Although there is a robust culture of manager (often a contractor) must recruit usually kept in a separate EDC system—
independence among designers of and train investigators to undertake the Finally, companies should redefine an or even on paper.
trials, several factors are encouraging trials. In most cases, the physicians who important factor in trials: the role of clinical-
standardization and reuse. The first is the act as investigators have to balance research associates. Currently, associates A single, real-time view of all the
recognition that integrated planning helps competing demands: the extra effort mainly monitor programs. Instead, they relevant data on trials could help
make better use of the organization’s required to carry out cutting-edge research
resources. Creating reusable modules with the need to devote sufficient time to A Sabre for pharmaceuticals?
that can be applied in a number of studies manage a practice and care for patients.
(for example, a standardized approach Conducting efficient clinical research Just as American Airlines’ Sabre system accelerated the adoption of electronic ticketing in the airline
to designing and collecting data about trials is therefore a primary concern industry by offering a standard booking interface across many air carriers, so too could a standard system
a drug’s safety and any adverse effects) for investigators. Any improvement speed up the adoption of electronic data capture in pharmaceutical trials. Although investigators and other
saves time in subsequent designs. Senior in efficiency will help pharmaceutical researchers currently have to learn a new system for every pharmaceutical company or clinical research
managers, recognizing this opportunity, companies to sign up leading practitioners. organization with which they work, change is on the way.
are often strong advocates of the shift to
modular design. In recent years, investigators have become One initiative, sponsored by the US National Institutes of Health, is called the Clinical Research Information
increasingly frustrated by EDC systems, Exchange. It aims to bring commercial and academic researchers together in a system that automates and
Second, the advent of EDC creates which, they say, create more work than the standardizes registration forms for trials. However, because industry-wide efforts often move slowly, other
an incentive for designers of trials to old-fashioned pen-and-paper method did; organizational forms may emerge to drive the next level of standardization. An open-source foundation
modularize the process in order to sometimes, for the sake of reliability they could develop a core set of standard tools coupled with a for-profit ecosystem of service providers that would
minimize the cost of developing and have had to keep data both ways. Doctors supply, maintain, and configure such systems as part of their work with pharmaceutical companies. A small
integrating electronic case reports for and nurses also complain that they lack consortium of pharmaceutical companies, working in the same therapeutic areas, might coordinate their
each one. First-generation EDC systems adequate training in the systems—a efforts in electronic data capture. Finally, a commercial organization, financed by venture capital or private
created some unforeseen fixed costs. For problem complicated by the fact that every equity, could sign up large pharmaceutical companies as its lead customers to drive the standardization of
example, before they were introduced, company uses its own proprietary systems. systems across the industry.
38 Pharma R&D Compendium 2010 39
Using IT to speed up clinical trials

companies to overcome these barriers. as alerts to warn when data entry has
IT suppliers hope to provide more been delayed—is one example of how
integrated systems, but the task IT can help managers to coordinate
may take a while. In the meantime, projects to ensure that bottlenecks do
therefore, companies should continue not occur. Alerts could also help to trigger
to take a best-of-breed approach and coordination across functions to keep
invest in middleware tools that help processes moving without delays.
to exchange data among systems. A
“dashboard” that displays key metrics Finally, a standard interface can help
in a spreadsheet or some other easy- companies to analyze and compile data
to-view form can also help managers to received from a number of different
keep tabs on the bigger picture. partners that manage studies within a
single program. Currently, each third
Even across a number of systems, party might use its own data standard.
companies must work to streamline Standardizing within a company (and
and unify the way they manage work across the industry) could make it easier
flows. Automated notification—such to exchange data and speed up trials.

Sam Marwaha (sam_marwaha@mckinsey.com) is a director in McKinsey’s New York office, where


Navjot Singh (navjot_singh@mckinsey.com) is a principal. Samir Patil is a McKinsey alumnus.

1
Bayes’ theorem explains how to update or revise beliefs in the light of new evidence.
2
Enterprise project-management platforms are large project-management systems that enable
companies to manage portfolios of projects more efficiently across a whole organization.
Depending on a company’s needs and capabilities, such a system might be either a custom
package developed by a single vendor or a combination of “best-of-breed” applications and tools.
3
At many pharmaceutical companies, systems for developing trials are fragmented into five clusters:
systems to manage trials, to capture data, to manage data safely, to manage documents, and to
manage projects and resources.
Pharma R&D Compendium 2010 41
Quality gates: raising the standard of pharmaceutical technical development

Good technical development can improve quality, lower


Quality gates: raising the costs, and get products to market more quickly, yet few
standard of pharmaceutical pharmaceutical companies manage it well. Quality gates
can transform the process.
technical development
Michele Holcomb, Martin Møller, and Paul Rutten

New drugs have to be clinically effective. Innovations in technical development


But to be commercially successful, can also open up new therapeutic uses,
they also have to be mass produced extend the life of a patent, and help
safely and reliably in a form that products win a distinctive market position.
ensures a consistent therapeutic effect. Yet many companies still struggle to
This technical-development process1 manage their technical-development
absorbs between 15 and 30 percent processes well. Indeed, we believe that
of the development costs of a typical the pharmaceutical industry could raise its
small-molecule drug and even more profits by up to 20 percent if companies
in biologicals. Indirectly, it affects 50 systematically pursued opportunities
percent of those costs if one includes, to reduce the costs and increase the
say, delivery of the supplies needed to run revenue of technical development. A
clinical and non-clinical trials.2 management tool known as the quality
gate is key to reaping such rewards.
Good technical development can
improve the quality of products and Identifying risk early
manufacturing processes,3 lower costs,
and get products to market more quickly. Technical development is a complex and
cross-functional activity
EXHIBIT 1 that must overcome
Quality gates in the development cycle manufacturing challenges,
Phase I Phase II Phase III
First full-
scale
take account of quality
FPFV1 FPFV POP2 FPFV batch File Launch
assurance, intellectual
Time
property protection, and
1 2 3 regulatory compliance,
Ready to Ready for Ready for commercial
start Phase II scale-up production and address scientific
▪ ▪ Choice of final API synthesis and formulation and ▪ Readiness for validation of
Forward-looking project
plan including resources packaging processes, including analytical methods the drug product process and medical issues. The
▪ ▪ Approval of preliminary manufacturing process ▪ Risk assessment

Risk assessment
Formulation choice for design ▪ Completeness of traditional management
Phase II3 ▪ Risk assessment technology transfer
▪ Choice of preliminary API4 ▪ Acceptability of estimated product COGS5 ▪ Readiness of sites for approach is reactive:
synthesis route ▪ Approval of forward-looking process-development inspections by authorities
▪ Acceptability of API cost and technology-transfer plan ▪ Consistency of the full- when problems occur,
▪ Need for additional API ▪ Choice of manufacturing sites, commercial scale scale process with original
process research suppliers, and testing sites filing cross-functional groups
▪ Readiness to produce ▪ Readiness to produce Phase III supplies
Phase II supplies ▪ Readiness to file, in accordance with submission
strategy
come together to find
solutions. But the end
result is often higher costs
1
First patient first visit.
2
Proof of principal.
3
All choices assumed to include the relevant analytical methods.
4
Active pharmaceutical ingredient. or launch delays.
5
Cost of goods sold.
42 Pharma R&D Compendium 2010 43
Quality gates: raising the standard of pharmaceutical technical development

Part of the problem has been that development project against a clear EXHIBIT 3 The process should be
R&D managers give scant attention to target state with attention not only to Traffic light coding easy to follow and to
technical development, often regarding development speed, but also to future communicate. Evaluation
it as an enabler rather than a driver cost (or process robustness) and potential Evaluation Definition Consequences and target criteria should be
of value creation and focusing solely quality or regulatory issues. Any deviations clear and readily understood.
▪ There are significant gaps in ▪ Corrective action defined
on development speed. Even when are identified quickly and appropriate the work OR
▪ Significant deviations from
▪ Necessary escalation taken in
a timely manner
managers do try to assess technical remedial action is agreed. Importantly for targets are expected OR
▪ Major risks exist which cannot
3. They should be
▪ Strict follow-up defined

development, the highly specialized pressed managers, assessing a project be mitigated


consistent and predictable.
and science-based nature of the work at a quality gate is not a lengthy process. ▪ Current status deviates
significantly from targets, but
▪ Technical development and
clinical project leadership
Gates should use the
makes evaluation difficult. The first A typical quality gate review takes less actions are in place to close
the gap OR
acknowledge deviations, risks,
and action plan same evaluation criteria for
▪ There are significant risks, but ▪ Arms-length monitoring of
indication of trouble often comes only than half a day, and some companies actions are in place that progress defined all projects and progress
mitigate the risks
when the technical-development team use as few as four gates during the should be tracked
misses a critical project milestone. clinical-development cycle of a new drug ▪ No/little deviation from target
status at current time
▪ No corrective action needed throughout the project
to assess whether it is ready for clinical ▪ Limited risks using consistent metrics.
Some companies are starting to do trials, ready to start Phase II, ready for In this way, information
things differently. Quality by Design is scale-up, and ready for commercial is easily accessible,
a proactive, risk-based approach to production (Exhibit 1). comparisons can be
technical development that helps identify made between projects,
potential problems before they occur, How quality gates work quality, and costs. Meetings should be and those who perform the quality-gate
and puts mitigation strategies in place designed using three basic principles. reviews gradually develop a valuable sense
to ensure that the interests of all relevant Quality-gate meetings are attended of pattern recognition.
functions are served quickly, smoothly, by key members of the project team 1. They should be purposeful and proactive.
and cost effectively. At the heart of and a management committee, often This means they should add value for Quality gates call for a collaborative
this new approach are quality gates, consisting of functional heads of chemistry, all participants: they are not a reporting working style, and participants must
used extensively in industries such as manufacturing, and controls (CMC), exercise. The objective is to smooth commit themselves to raising any
the automotive sector to manage the production, quality assurance, and progress towards project goals, both in the problem as soon as possible. Discussions
complex, costly, and time-consuming regulatory affairs, and preferably also a near and longer term. should be open and constructive,
development process, and applied with senior business leader such as head of concentrate on finding solutions quickly,
great success in clinical development R&D. All functions need to be represented 2. They should be simple. Each gate should and culminate in defining next steps,
in the pharmaceutical industry. Many to ensure the meeting focuses on time, require the minimum necessary preparation. individual responsibilities, and deadlines.
pharmaceutical companies
claim to be using quality EXHIBIT 2 EXHIBIT 4 Measuring progress
gates to manage technical Evaluating progress Warning signs
development too, but At the heart of the quality-
Process design
few do so rigorously Evaluation criteria Quality gate 2 – ready for scale-up gate process is a set
Process Process realization API Formulation Packaging
enough to reap the maturity
1. Process properties of metrics that evaluate
Process readiness
potential rewards. Used 1.1 Process
design
1.1.1 Process options Final API synthesis route Final formulation has
has been defined been defined. Equipment
Final packaging process
has been defined.
every aspect of technical
well, quality gates can Product
Product design train for production has
been chosen
Equipment train for
production has been development—the process,
maturity chosen
transform the technical- Product performance
For every quality gate, key
1.1.2 Value-chain Full value-chain map Full value-chain map Full value-chain map
product, costs, legal, and
criteria remain the same.
development process. Evaluation
criteria Cost and quality
Product-cost status However, targets are set at
map includes yield rates,
process times, and all
includes yield rates,
process times, and all
includes yield rates,
process times, and all
regulatory status, and CMC
different levels for each
optimization
Quality by design review
secondaries (for
example, solvents)
secondaries (for
example, solvents)
secondaries
status (Exhibit 2). These
Quality gates provide a Readiness for filing
1.1.3 Difficulty
assessment
List of difficulties exists,
plan to handle is spelled
List of difficulties exists,
plan to handle is spelled
List of difficulties exists,
plan to handle is spelled
are defined as “maturity
mechanism to enforce Regulatory/legal
Freedom to operate
out (QA, HSE, supplier
capabilities, etc)
out (QA, HSE, supplier
capabilities, etc)
out (QA, HSE, supplier
capabilities, etc) metrics,” their purpose
regular, cross-functional 1.1.4 Starting material Starting materials Starting materials
being to measure progress
Risk assessment
evaluation of development CMC1 status
choice including GMP/non-GMP
status have been
including GMP/non-GMP
status have been
towards developing and
Schedule adherence
risks. They measure the confirmed confirmed
manufacturing a low-cost,
progress of the technical- high-quality product that
1
Chemistry, manufacturing, and controls.
44 Pharma R&D Compendium 2010 45
Quality gates: raising the standard of pharmaceutical technical development

FICTITIOUS EXAMPLE
is delivered on time. Each EXHIBIT 5 start addressing problems they uncover comfortable with collecting and sharing
function is responsible The full picture straightaway. Individual, preparatory team data that gives a realistic overview of
Example problem areas

for evaluating progress in Evaluation criteria


Quality gate 2: ready for scale-up
API1 Formulation Packaging
meetings ahead of a quality gate are project progress. And, importantly,
its own area of expertise. 1. Process properties

Information in API value-


therefore important. companies must raise the profile of
1.1 Process design
However, given the cross- chain map has major gaps
technical development so that its value-
1.2 Process realization Selection of production
functional nature of the 1.3 Process readiness
facilities for API and drug Some companies find it helpful to form a adding role is recognized.
product critically delayed
work, other functions in the 2. Product properties dedicated technical-development project
2.1 Process design
quality-gate team need to 2.2 Process
office to provide logistical support for the Pilot schemes will help. By using quality
performance
endorse each evaluation. 3. Cost and quality optimization quality-gate process. Ultimately, however, gates on one or two key projects with
3.1 Product cost status Gaps in cost-reduction
activity and design-space the success of quality gates depends extensive management and administrative
application, specifically in
A smart mix of qualitative 3.2 Quality design
formulation upon those involved adopting a forward- support, companies can fine-tune the
4. Regulatory, legal
and quantitative metrics 4.1 Readiness for filing Dossier text not reviewed
looking, risk-based way of thinking process and successfully demonstrate the
can effectively evaluate 4.2 Freedom to operate
5. Overall CMC status 2
cross-functionally
about their work. Participants need to be value of the new way of working.
FMEA in formulation still 3

progress against defined 5.1 Risk assessment


5.2 Schedule
points out too many risks
adherence
targets without greatly 1 Michele Holcomb (michele_holcomb@mckinsey.com) is a principal in McKinsey’s New Jersey office.
Active pharmaceutical ingredient.
increasing the workload Chemistry, manufacturing, and controls.
2

Failure modes and effects analysis.


3
Martin Møller (martin_moller@mckinsey.com) is a principal in the Copenhagen office. Paul Rutten
of the teams involved. So, (paul_rutten@mckinsey.com) is an associate principal in the Amsterdam office.
for example, a quantitative
measure of process-design maturity, also include an evaluation of the risks
itself a sub-category of process maturity, involved in taking the project forward
would be the number of impurities in the to the next stage, and a perspective
formulation. Qualitative metrics are needed on how the manufacturing process
for areas that are less easy to define, will evolve towards commercial
such as “freedom to operate,” that is, production standards. Thus, the quality
whether a product infringes any patents. gate encourages agreement about
what needs to be done to fix existing
The metrics are the same at each problems and to reduce the likelihood of
quality gate, though the targets are future problems.
adjusted to reflect the different phases of
development. A traffic light color-coding The evaluations from all the various
system then indicates the extent to which project areas are finally combined in a
the project is on target, again using single chart, giving a clear overview of
standardized definitions (Exhibit 3). all aspects of the technical-development
process, and showing at a glance
This coding system is applied to every where risks and problems lie and
sub-category of the evaluation criteria. decisions need to be made. The chart
Exhibit 4 illustrates this for process effectively dictates the agenda for the
design, showing the current status of quality-gate meeting (Exhibit 5).
the active pharmaceutical ingredient
(API), formulation, and packaging Part of the value of the quality gate is
against certain targets. The warning the opportunity it gives participants to
signs here are that there is no map learn from the experiences of others on
of the API value chain and solvent different projects. However, much of
1
The process has various other names, including chemistry, manufacturing, and controls (CMC);
usage has not been analyzed. the value of the quality-gate process is CMC development; process research; pharm sci; and chemical pharmaceutical development.
often realized before the meeting. In the
2
Ted Fuhr, Michele Holcomb, and Paul Rutten, “Why ‘Quality by Design’ should be on the pharma
Importantly, the metrics do not only course of collecting the data needed for CEO’s agenda,” http://operations-extranet.mckinsey.com.
record project progress to date, they the quality gate, individual teams often
3
Anil G. D’souza, David J. Keeling, and Richard D. Phillips, “Improving quality in pharma
manufacturing,” mckinseyquarterly.com, September 2007.
Pharma R&D Compendium 2010 47
Market-access-minded development

A new way of conceiving clinical development will


Market-access-minded development uncover the drugs bound for commercial success
early on. Think value.

Matthias Evers

Recent research shows that clinical It will be more value-focused and forward
differentiation against the standard of care looking. Delivering a specified efficacy
is the best indicator of a drug’s future after four weeks of treatment might be the
commercial success.1 This finding is borne right endpoint to choose as far as winning
out by the high-profile failures of products approval for a drug is concerned. But if
identified as being insufficiently differentiated the specification does not differentiate the
or as cost-inefficient by institutions such as drug against the standard of care at the
Germany’s Institute for Quality and Efficiency time of launch, it will not help to make it
in Health Care and the UK’s National successful commercially. Clinicians and
Institute for Clinical Excellence, and by their teams thus need to move beyond
managed-care organizations. To win market the “target product profile” to think
acceptance as well as regulatory approval, through how the unmet needs of patients,
pharmaceutical companies must develop physicians, and other stakeholders can
drugs that are differentiated and conduct be met, and to design trials that reveal
trials for them in a way that demonstrates what added value the drug can deliver.
their value convincingly. It is therefore important not to tweak the
profile to suit the clinical data, but regularly
We have coined the term “market-access- to check that it still reflects stakeholders’
minded development” to describe a new needs. Because these needs evolve, and
way of crafting clinical strategies and are likely to differ by region and perhaps
designing trials. The crucial difference is that, country, a clear definition of desired patient
as early as the beginning of Phase II, clinical outcomes and cost effectiveness needs
teams start thinking beyond the endpoints, to be part of the traditional approach to
patient segments, and study designs that building a target product profile.
line the path to approval, and focus on
value as well. Not all stakeholders will define It will be more targeted and front-loaded.
value in quite the same way, but most seek Clinical teams will apply patient outcomes,
evidence of better patient outcomes. cost-efficiency and economic milestones
early in development, that is, before
Characteristics of this they initiate more expensive late-stage
new approach to development Phase III trials. Just as “key opinion
leaders” from science are currently
Market-access-minded clinical development involved early in development, so key
is likely to include some or all of the opinion leaders from the realm of health
following four characteristics. economics (such as health economists
48 Pharma R&D Compendium 2010 49
Market-access-minded development

from academic institutions) will be development and clinical teams must on new designs), functional silos, and what needs to change in order to shift
brought in to assess and communicate think beyond the underlying scientific a poor interface between development their company’s approach from a focus on
the cost effectiveness of a new drug. Their mechanisms and consider how to and commercial. Clinicians’ reluctance approval to a focus on patient and broader
expertise, as well as the clinical team’s generate early insights into the way the to accept that cross-functional efforts stakeholder value. Cultural change is likely
creativity, will deliver innovative clinical compound might address patient needs. are now essential, and a shortage of to be necessary, as are new capabilities.
strategies that characterize a drug’s Armed with such insights, the clinical capabilities in emerging disciplines such
potential value to patients during proof- strategy can be crafted to focus on as health economics, outcomes research, Convincing in-house success stories help
of-concept trials and in Phase II. value in a single indication and across and the gathering of payors’ views, are to break the mould. We urge managers
indications. Execution plans for the likely further inhibiting factors. to start by conducting pilots with selected
It will be more rigorous. Instead of relying more extensive Phase II trial programs will (early) project teams. When high-profile
on a tiny group of senior experts who also focus on value, while simultaneously There are no instant solutions: teams embrace market access as an
“just know” from experience what trial maximizing speed and mitigating risks. development leaders need to think additional challenge and demonstrate the
designs to pursue, market-access-minded Though this might lead to higher costs systematically about all the functions and benefits of the new approach, it becomes
clinical development is more rigorous and and attrition in Phase II, it should facilitate departments at the interface between easier to initiate more fundamental and
systematic. It is structured in four steps: the demonstration of better value in development and commercial, and ask widespread change.
define which of a drug’s claims are the Phase III, yielding data that can be used
most valuable to prove, identify the most immediately to negotiate and achieve Matthias Evers (matthias_evers@mckinsey.com) is a principal in McKinsey’s Hamburg office.
suitable patients, define the most relevant market access.
endpoints, and deploy the most cost- The author would like to thank Valentina Sartori, Jerrina Eteen, and all other colleagues involved in
efficient design. Obstacles which McKinsey’s Value-Driven Drug-Development work.
tend to get in the way
It will be more cross-functional. Clinical
strategy and trial design are often It is challenging to transform the way
considered the home turf of the senior pharmaceutical companies develop
brand clinician. In the new world, clinical drugs, shifting their focus away from
strategy and trial design will become a achieving approvals to creating value
more cross-functional effort involving a and marketable labels. One impediment
broader group of experts: pricing and is the scarcity of people who excel at
reimbursement specialists to provide the clinical strategy and trial design. Even
payor’s point of view, health economics leading companies rely on a very small
and outcomes researchers to advise on group of experts whose skills are based
the endpoints that demonstrate cost on experience and pattern recognition.
effectiveness, marketing experts to give So far, their abilities have not been
the customer’s perspective, modeling and codified into a rigorous approach that
simulation specialists and biostatisticians would support less experienced co-
to indicate the most suitable patients and workers. There is little or no training to
cost-efficient designs. The composition, turn potential into proficiency, and too
modus operandi, and performance much reliance on the copying and pasting
management of the clinical-project of supposedly proven designs instead of
team as well as individual job profiles learning from mistakes and innovating.
will increasingly need to guarantee such
cross-functionality. Other barriers are cultural and
organizational. They include an ingrained
In summary, market-access-minded habit of focusing on clinical parameters
development is value-driven. Right from rather than value, risk aversion (it is easier
the outset, when clinical trials are set to replicate trials with endpoints that have
up to demonstrate proof of concept, led to registration in the past than to work
Maria Gordian, Navjot Singh, and Rodney Zemmel, “Why products fail in Phase III,” In Vivo, April 2006.
1
Pharma R&D Compendium 2010 51
Changing with the times: how pharmaceutical companies should respond to regulatory change

Drug companies need to make organizational changes to meet the


Changing with the times: how latest patient safety regulations in the United States. But as the
pharmaceutical companies should regulations evolve, companies will also need to work openly with all
stakeholders to ensure the right outcome.
respond to regulatory change
Maria Gordian and Richa Pande

In response to concerns about patient needed to reach the right regulatory


safety, pharmaceutical risk-management outcome. Here, pharmaceutical
programs in the United States have companies have a key role to play.
become increasingly stringent. Managing as they do the conception,
Since March 2008, the Food and Drug development, testing, and launch of
Administration (FDA) has required new treatments, they have access to
manufacturers to submit a risk evaluation data that will be invaluable in efforts
and mitigation strategy (REMS) to manage to improve risk management, helping
known or potential safety risks related to all stakeholders to make objective
certain drugs based on available safety decisions about the safety of medicines
data,1 and thus to help ensure a drug’s and the risk/benefit trade-offs.
benefits outweigh its risks. As a result,
pharmaceutical companies need to make A brief history of
changes across their organizations, paying pharmaceutical risk
more attention to safety at each stage management
of drug development and building new
organizational capabilities. Regulatory risk-management
programs are by no means new.
But the regulatory environment is However, the trend has been towards
still evolving. Although the FDA has broader and tighter regulation.
established a broad framework for risk
management, many details have yet to In 2004 and 2005, more than 20 drugs
be settled. It is still unclear, for example, were withdrawn from the US and
how the level of risk is decided. And larger European markets following a wave of
issues, such as how best to measure injuries and deaths that were blamed on
the impact of a REMS, have not been new prescription drugs. Many lawsuits
addressed. Moreover, while patient safety ensued, and public officials, including
remains the paramount goal, it is important legislators and the FDA, came under
not to overburden the health care system, intense public pressure to place more
or to prevent some patients from receiving emphasis on drug safety.
potentially life-saving drugs.
Prior to 2005, risk-management programs
Input from all stakeholders— were implemented on an ad hoc basis.
pharmaceutical companies, pharmacists, Mitigation consisted largely of education
health care providers, and patients—is programs and changes to product
52 Pharma R&D Compendium 2010 53
Changing with the times: how pharmaceutical companies should respond to regulatory change

labeling, although occasionally distribution (NMEs) and novel biologics, and dozens For their part, pharmaceutical companies is recognized that the risks outweigh the
was restricted too. Clozaril’s “no blood, of marketed products, have received must now develop more sophisticated benefits. Furthermore, companies will be
no drug” program in 1990, for example, REMS requests from the FDA. Between programs to communicate with all those able to avoid situations whereby the FDA
required schizophrenia patients to register January and June 2009, however, about who prescribe, dispense, and administer imposes new trial requirements late in the
for blood tests to monitor the risk of 45 percent of the 38 new products a drug with REMS requirements, and development cycle, creating unforeseen
leukopenia and agranulocytosis. approved were required to have post- manage and monitor them too. But their delays and costs.
market commitments, indicating that the approach to risk management will have to
When studies revealed that the existing proportion of products requiring a REMS is extend far beyond this. Build efficient REMS-management
risk-mitigation programs were not effective, likely to rise. processes
the FDA took steps to standardize Organizational change Companies need to establish efficient
and improve them further, issuing its REMS requirements can have implications management processes not only to meet
guidance on risk-minimization action plans for everyone in the pharmaceutical value To operate efficiently and effectively in the FDA requirements promptly—they have
(RiskMAPs) in March 2005. RiskMAPs chain. The fact that certain medicines might new REMS environment, pharmaceutical 30 days to produce a medication guide,
consisted of three elements: targeted be available only through certified physicians companies will need to adopt new for example—but also to prevent any
education and outreach programs; and pharmacies could prove problematic decision-making and management costly launch delays. Although some
reminder systems, which were essentially for patients living in remote areas or with processes, adjust their organizational requirements can be satisfied only on a
more elaborate education programs; and limited access to the health care system. structures, and develop new capabilities case-by-case basis—new clinical trial
performance-linked access systems or Pharmacists might have to seek additional for managing risk from early development requirements, post-marketing studies,
restrictive-distribution programs. certification and provide more patient through to post-loss of exclusivity (LOE). or surveillance programs, for example—
education, and some might need to comply Specifically, they should: others, such as the writing of medication
Safety concerns and public and political with risk-management plans aimed at guides, can be met by standardized
pressure continued to mount, however, helping to prevent drug abuse and overdose. Anticipate and identify possible processes that will speed compliance,
and the FDA now imposes REMS Physicians who wish to prescribe certain safety risks, and communicate lower expenses, and improve safety.
requirements on those drugs it feels drugs might also need to obtain additional early with the FDA
warrant tighter safety assessment and certification, spend more time educating Companies should be able to forecast Incorporate safety, risk-evaluation,
management. According to one of its patients, and adhere to restrictions on which if their drugs will be subject to REMS and risk-mitigation considerations at
Senate sponsors, the REMS legislation patients can receive some treatments. Even requirements—there should be no every stage of a product’s life cycle
is intended “to ensure that drug safety is the FDA faces challenges, trying as it is surprises. Identifying potential safety risks While the FDA typically imposes REMS
not an afterthought but an integral part to meet its expanded responsibilities with early in development must become a requirements when a product is approved
of the process throughout the life of a limited resources and budget. habit, as must managing those risks. If a or when new safety data becomes
drug.” Exhibit 1 shows the drug seems to be causing elevated liver available for a product already on the
various potential REMS EXHIBIT 1 enzyme levels in early development, for market, risk evaluation and mitigation
requirements: an approved Potential REMS requirements example, every effort should be made to should inform key decisions throughout
medication guide; a collect the data that explains the problem the life cycle of a drug. Actively addressing
Description
communication plan for ▪ Leaflet for patients to inform them of the risks and benefits of the medication. Distributed by the and to share it with the regulator, not only safety concerns raised by any preclinical or
Medication guide pharmacy when a prescription is filled
health care providers; to demonstrate adequate diligence but clinical data should be a priority, and key
elements to assure safe ▪ Educational materials for health care providers to support REMS implementation, for example,
also to address properly any concerns development decisions (including go/no-
Communication
use, such as physician plan
letters, advertisements in trade journals, memos to professional societies
about the potential frequency and severity go choices, safety and efficacy endpoints,
training and certification, of safety events. By sharing data with and clinical trial design) should all be
▪ Active management of safety risks including:
and patient education; an Elements to
ensure safe use
– Prescriber education and certification
– Dispenser certification
regulators earlier and more frequently considered from a REMS perspective. A
implementation plan; and a – Restricted administration (for example, given only in hospitals)
– Dispensation to patients only with evidence of safe-use conditions
during development, companies will be company’s medical, science, regulatory,
timetable for assessment of (for example, laboratory test results)
– Patient monitoring able to help shape the understanding of legal, and commercial teams should all be
– Patient registry
the REMS. a drug’s risks and benefits, and whether involved. Exhibit 2 describes the triggers
▪ System to monitor, evaluate, and improve implementation of “Elements to ensure safe use”
Implementation
system
a REMS might be required. They might that should prompt team action. In early
Since implementation, even find that the targeted clinical profile development, for example, knowing that a

about 25 percent of Assessments
Reports to the FDA on the implementation and effectiveness of REMS at least 18 months, 3 years,
and 7 years post-REMS approval of the drug changes so that it can be mechanism of action has safety issues or
new molecular entities used in certain circumstances where it that other products in the same class have
54 Pharma R&D Compendium 2010 55
Changing with the times: how pharmaceutical companies should respond to regulatory change

REMS requirements should EXHIBIT 2 Thought also needs to be given to It is in these kinds of communications
trigger the collection of Safety triggers how safety can be improved without between all stakeholders that
more data. It should also overburdening the health care pharmaceutical companies have an
prompt discussions with Timing/phase of Early development Late development In-market Post-LOE
infrastructure. The FDA is sensitive to important role to play. With so much
regulators as early as product
development
Discovery and
preclinical testing
Clinical development
(Phases II and III)
Marketed/mature
products requiring
Products that have
lost market exclusivity
practitioners’ concerns about having to data at their fingertips, they are an
Clinical development
Phase II or III to make NDA submission
comply with different companies’ REMS essential participant in discussions aimed
1
REMS (reactive
(Phase I) NDA review versus proactive)

sure there is agreement for drugs in the same class, and so at improving the safety of medicines,
on what data might be has been considering whether a single, understanding the risk/benefit trade-
required to establish Example
triggers
▪ Mechanism of action
with known safety
▪ FDA requirement for
product in response to
▪ FDA requirement for
product in response
▪ FDA requirement for
product in response to
class-wide REMS might be appropriate offs, and managing those trade-offs.
the product’s safety issues data/submission to data/submission data/submission
for extended-release opiods. To help it in Pharmaceutical companies can and
▪ Class labeling/REMS ▪ Class labeling/REMS ▪ Class labeling/REMS ▪ Class labeling/REMS
profile. Similarly, new ▪ Safety issues ▪ Safety issues identified ▪ New safety signals ▪ New safety signals
its deliberations, it has called for a series should take the initiative whenever
safety signals as a result identified through
preclinical, toxicity,
through Phase II or
Phase III clinical data
of meetings with different stakeholders possible to facilitate the open exchange
of pharmacovigilance and Phase I data
to discuss the possible implications, of information, actively participating
or post-marketing including how competitors might work in debate and, when appropriate,
studies might prompt a together to develop a common REMS; themselves bringing together stakeholders
REMS requirement at New drug application.
1 the IT infrastructure required to support by convening round-table or working
extremely short notice, a REMS that would cover more than sessions. The aim is to establish
so companies need to be 20 million prescriptions a year; how its a transparent process whereby all
prepared to meet tight deadlines and need the capabilities required for risk effectiveness would be measured; and stakeholders work towards shaping the
avoid civil penalties. management, from product development who should be responsible for operating right regulatory outcome—an effective
to commercialization. the REMS program. approach to ensuring patient safety.
Understand the financial implications
A REMS will affect budgeting decisions Regulatory work in progress Maria Gordian (maria_gordian@mckinsey.com) is a principal in McKinsey’s New York office and
and commercial value. Companies Richa Pande (richa_pande@mckinsey.com) is a consultant in the New Jersey office.
therefore need to factor it into The new REMS regime is still work in
their revenue forecasting, product- progress. Operational aspects are being
development investments, and the worked out as the FDA and industry
costs related to product launch, sales, together decide the best way to meet
marketing, and post-marketing. REMS- REMS requirements. Companies are
related costs and revenue implications currently required to lay out detailed plans,
also need to be considered when valuing which the FDA will then approve. But with
assets for portfolio prioritization, and for time and experience, it is likely that there
licensing and acquisition purposes. will be greater clarity and guidance on
what constitutes best practice.
Develop new capabilities
Most companies will need to acquire For the time being, there are many
new capabilities so that they can unanswered questions. How, for
respond quickly to and manage REMS example, can the success of a REMS be
requirements, mine internal and external measured? Is it feasible to collect and
data more thoroughly for earlier warning analyze data in a way that will reveal what
signs about risks, and continuously works and what does not and to modify
monitor and assess risk-management programs accordingly? And how should
programs both for reporting purposes “success” be defined if restricting access
and to improve them. While the delays to certain medicines for safety reasons
and costs associated with these efforts denies life-saving treatments to patients
might be particularly onerous for prepared to make a different cost/benefit
smaller companies, every company will trade-off?
Their own data or that available for similar drugs.
1
Pharma R&D Compendium 2010 57
The anatomy of attrition

Research tracking the attrition rates of more than 3,000


The anatomy of attrition compounds in each phase of development reveals recent
drug failure trends.

Navjot Singh, Rodney Zemmel, Maria Gordian, and Áine Denari

More than 90 percent of compounds that To update our research, we conducted


enter Phase I trials are destined to fall out an outside-in analysis of pharmaceutical
of the development pipeline, making drug R&D attrition rates between 1996 and
failure a headline issue for the pharmaceutical 2007. Using publicly available data, we
industry, academics, analysts, and drug tracked each phase of more than 3,000
regulatory authorities. Attrition rates of this compounds in development (see sidebar,
magnitude are the single biggest cause of “Methodology”). By tracking phases rather
the industry’s R&D productivity challenge. than full projects, we were able to capture
what has been happening quite recently in
Many companies have made concerted the industry, and so assess trends in more
efforts to cut attrition rates (see sidebar, detail. Here are the main findings.
“Cutting the losses”), and much research has
been conducted to explain why it continues Overall clinical success rates have
to be so high. In 2006, for example, worsened in the past 12 years
McKinsey research showed that almost half Across the industry, the success rate
of all failures in Phase III were due to a lack of of drugs fell between 1996 and 2007,
demonstrable efficacy of a drug compared and worsened in the second half of that
with a placebo. period. Almost all major biopharmaceutical
companies followed the
EXHIBIT 1 trend (Exhibits 1 and 2).
Overall decline
Across the industry, the success rate of drugs has fallen, mainly because of the rising level of attrition in Phase II
Higher attrition in Phase
3-year rolling average rates, % II is the main reason for
70 declining success
65 Phase I
Phase III
As the exhibit shows,
60
55 attrition during Phase I
50 has been relatively stable.
45
40
After a dip earlier in the
35 Phase II analysis period, success
30
25
rates in Phase III are now
20 recovering, leaving attrition
15
10
Phase I–Launch1 in Phase II as the single
5 biggest reason for declining
97 98 99 00 01 02 03 04 05 06 07
success. Outside-in
1
Phase I-Launch success rate calculated as (Phase I 3-year rolling average x Phase II 3-year rolling average x Phase III 3-
year rolling average x 90%); using 90% assumption for regulatory/launch success. analysis suggests a 16
Source: McKinsey R&D Attrition Database (excluding formulations); Pharmaprojects; McKinsey analysis
58 Pharma R&D Compendium 2010 59
The anatomy of attrition

Cutting the losses

The approaches that companies are using to reduce attrition rates fall into four main categories: Finding alternative uses for drugs
Ultimately, of course, launching a drug is more valuable than stopping one that is likely to fail. It is vital
A better understanding of the causes of failure therefore to explore alternative indications for a candidate drug that fails in the primary indication, as long
Companies have invested in developing internal databases that track in great detail the history of compounds, as the exploration is justified. Many companies have tried to systematize searches for alternative indications.
pinpointing when they failed and why. These databases have proved effective in helping companies understand Some, for example, have created groups that look for common pathways in the discovery phase, others focus
the root causes of failure and to take corrective action—for example, by changing milestone-driven guidelines on clinical investigations.
and introducing new assays. This input also provides useful feedback to discovery scientists who design new
molecules. Modifying portfolio strategies
The choices that companies make in relation to their portfolios are one of the most important ways of reducing
Earlier identification of drugs likely to fail attrition. There are four that companies typically consider.
Scientists in the biopharmaceutical industry understand that attrition is part of development. The primary
strategy of most biopharmaceutical companies has therefore been to identify at an early stage those drugs that The balance of proven and unproven mechanisms in the portfolio. Even though the expansion of target
are likely to fail, and to stop their development and avoid wasting money. There are three ways companies are space in the clinic has been minimal, the attrition rate of unproven mechanisms is almost twice that of proven
pursuing this. mechanisms. What ultimately matters is meaningful clinical differentiation. How best to balance proven and
unproven mechanisms in the portfolio remains a much-debated topic, and different companies have arrived
First, companies are investing in more small-scale in vivo and in vitro testing in late discovery to screen out at different conclusions.
compounds, especially those that might have toxicity issues. By contrast, efficacy biomarkers have been
moderately successful because of difficulties in human predictability. The number and diversity of back-up candidates. Advances in high-throughput screening have made it easier
to identify hits, but decisions about the diversity of hits, and the timing of the back-up, are critical and can have
Second, companies are increasingly pursuing high-throughput, early clinical-screening strategies to enable significant impact on overall program survival.
faster and more efficient clinical screening to test for efficacy in patients. These techniques include adaptive
clinical-trial designs and early patient testing, and the building of infrastructure to enable faster patient How to balance spending on life-cycle management and label expansion against investments in new molecular
screening. entities. Whilst the exact balance will differ by company, many have increasingly focused
on life-cycle management and label expansion in response to increased regulatory stringency.
Third, companies have enhanced their governance models and team structures. Over the years, governance has
become the focus of efforts to improve the survival rates of candidate compounds. Companies have improved Whether to move move away from the industrialized model for R&D. This model linked a set annual
their decision-making processes, governance philosophies, governance architecture, and the tactics and number of drug candidates with incentives and bonuses. The alternatives are a new model of proof of concept,
operations of governance. or a model that culls the number of preclinical candidates in the hope that only those with the best chances of
success will be advanced.

percent decrease in Phase II survival industry, as illustrated in Exhibit 3, and EXHIBIT 2


since 1997, leading to a survival rate remains a significant opportunity in terms Recent deterioration
Success rate Phase I-Launch,1 %
of 33 percent in 2007. Internal analysis of reducing attrition rates—failures due
of some companies suggests a Phase to lack of efficacy in Phase III should be
II survival rate of between 15 and 20 negligible if Phase I and Phase II trials are Company 1996–2001 2002–07 Delta

percent, particularly when survival rates conducted well. A


B
19.9 8.9 -11.0
20.7 19.0 -1.7
are assessed by indications (although C 13.9 11.6 -2.3

indication attrition is not always fully However, further disaggregation of D


E
15.6
20.5 8.1
17.1
-12.4
1.5

reported publicly). failures across five-year periods reveals F 18.0 10.1 -7.9
G 13.0 5.6 -7.4
three more interesting findings (Exhibit H 24.1 6.6 -17.5

Causes of Phase III failures are shifting 4). First, the number of failures due to I 30.5 15.4 -15.1
J 11.6 10.7 -0.8
Our previous research into drug failures lack of efficacy compared with a placebo Top 10 17.9 11.6 -6.3

in Phase III1 identified lack of efficacy is falling. Second, despite a number of Top 11-25 16.7 12.7 -4.0
Industry 14.8 10.3 -4.5
compared with a placebo as the main high-profile Phase III failures owing to
reason. This continues to be the single safety concerns, the relative fraction
1
Product of phase attrition rates that year; using 90% assumption for regulatory/launch success; for all
biggest cause of failure across the of such failures as a percentage of the originated and partnered drugs.
Source: Pharmaprojects; McKinsey analysis (excluding formulations)
60 Pharma R&D Compendium 2010 61
The anatomy of attrition

total has remained stable. EXHIBIT 3 EXHIBIT 5 with almost 150 percent
However, it is worth noting Causes of failure in Phase III Shrinking gap higher 12 years ago. The
Dramatic decline in the relative success of partnered and in-licensed compounds Organic
that the number of failures Partnered2 biggest pharmaceutical
1990 – 2007
in Phase III arising from % of overall failures
(n1 = 106) Success rates Phase I-Launch
companies seem to
Root cause Description
safety concerns that confirm %, 1996 to 2007
achieve similar results in
Efficacy versus placebo ▪ Failed to demonstrate efficacy
doubts raised earlier in compared with a placebo 45 33.8 both categories—a quite
development has increased. 28.5 different story from 12
Safety Confirmation of ▪ Safety issues raised in earlier trials or 25.8
This could imply that the versus early safety seen in similar class of on-market 8 +149% years ago, when their
placebo concerns compounds
industry is getting better 27 17.4 partnered compounds
Unclassifiable ▪ Unable to determine cause of safety +38%
at spotting potential risks failure from outside-in 19
13.6
10.5 11.4
9.8
13.3 13.1
had a 72 percent higher
in earlier phases, but that 6.5 success rate than their
Lack of Efficacy ▪ Given similar safety profile, failed to 9.5
it fails fully to evaluate the differentiation demonstrate superior efficacy 24 organic compounds. The
compared with active comparator
safety-risk benefits, leading 28 1996–97 98–99 2000–01 02–03 04–05 06–07 shrinking gap no doubt
Safety ▪ Given similar efficacy, failed to
to more expensive failures demonstrate superior safety 4 partly reflects the efforts
compared with active comparator
later in development. 1
For all therapeutic areas except formulation; using 90% assumption for regulatory/launch success.
made by many companies
1
Number of drugs in sample.
Source: EvaluatePharma; Pharmaprojects; Factiva; literature search; McKinsey analysis
2
Partnered compounds include those with a partnership of any sort during the given phase of development.
Source: Pharmaprojects; McKInsey analysis
to manage their internal
Third, the number of portfolios more rigorously.
failures due to lack of
commercial differentiation has risen slightly A dramatic decline in the
during the past five years—an increase relative success of partnered
probably driven by the higher demands of and in-licensed compounds
payors and access agencies. However, the Compounds developed in partnerships
question remains whether these failures were once the stars of drug development,
could have been avoided by Phase II trials far exceeding the success rates of
that better reflected a clear understanding those originated internally (Exhibit 5).
of customer needs. Although partnered compounds still
outperform “organic” compounds, the
Success rates have fallen gap has shrunk. Now, the success rate
in most therapeutic areas of partnered projects is 38 percent higher
Although the sample size is relatively than that of organic ones, compared
small because of the
limitations on establishing EXHIBIT 4
root causes of failures from Shifting patterns
Efficacy failures are decreasing, while differentiation failures are rising Primary driver
the outside-in, it would Secondary driver
Phase III failures, %
appear that success rates
in most therapeutic areas 1993 – 1997 1998 – 2002 2003 – 2007
(n = 22) 1
(n = 34) (n = 39)
(TAs) have declined. The
Efficacy versus
average decline is 4.5 placebo
45 53 44

percent (Exhibit 4), but the


Safety -
figure varies by TA. For confirmation
2 1 18

example, oncology success Safety -


27 22 8
rates have dropped by 3.2 unclassifiable

percent and endocrine Differentiation


18 24 27
by 13 percent. Only efficacy

cardiovascular success Differentiation


7 0 4
safety
rates have risen, and these
by just 3 percent. Number of drugs in sample.
1

Source: EvaluatePharma; Pharmaprojects; Factiva; literature search; McKinsey analysis


62 Pharma R&D Compendium 2010 63
The anatomy of attrition

Methodology

We conducted an outside-in analysis of pharmaceutical R&D attrition rates in the past 12 years,
tracking the phases of more than 3,000 compounds in development between 1996 and 2007. Our
research concentrated on orally ingested, small-molecule drugs (excluding biologics such as vaccines)
produced by large pharmaceutical companies, using Pharmaprojects data rather than companies’ self-
reported data.

We defined a drug as having “failed” if the trial ended early (but excluding those that ended early
because of strongly positive results), or if the trial failed to produce the results that would ensure
drug approval. We analyzed the cause of failure for those drugs for which there was sufficient public
information available, considering their efficacy and safety (as compared with placebos), and
comparing them with similar drugs already on the market. Our Phase III analysis consisted of outside-
in investigations to assess the causes of failure: analyst reports, press releases, and other sources of
public information. A panel of clinical experts reviewed our material to ensure we had interpreted
the data correctly. Though this data set is limited, we believe it to be more robust than the internal
information on trial failures available at any single pharmaceutical company.

The analysis only begins tracking phase success of a compound after the compound enters
Pharmaprojects, rather than including it as a success in earlier phases. This avoids over-counting the
number of successes and undercounting the number of failures in earlier phases. Each phase of each
project was tracked and analyzed independently based on the year it ended that particular phase (the
phase “exit year”). By tracking phases rather than full projects, we were able to look at composite
attrition numbers for the industry that reflect what has been happening quite recently. This allows for
a more detailed assessment of trends over time than can be achieved by other analytical approaches,
which tend to be limited in sample size.

The analysis highlights trends in R&D attrition rates, but also looks at the impact of other factors such
as therapeutic area (TA), company size, technology, and licensing status.

Historical in nature, the analysis makes no attempt to predict future trends, which are driven by
factors not easily modeled on the basis of historic data, including regulatory changes, portfolio
decisions, market access issues, and scientific evolution. Benchmarking at the TA level within individual
companies (or even entire small companies) is not always meaningful because of low sample size.

Navjot Singh (navjot_singh@mckinsey.com) and Maria Gordian (maria_gordian@mckinsey.com) are


principals in McKinsey’s New York office, where Rodney Zemmel (rodney_zemmel@mckinsey.com)
is a director. Áine Denari (aine_denari@mckinsey.com) is a consultant in McKinsey’s Dublin office.

The authors would like to thank Carla Zwaanstra, Susan Waters, Kendra Buzzell, Kenneth Yoon,
C.J. Pavia, Kristoffer Famm, and Tony Tramontin for their contributions.

1
Maria Gordian, Navjot Singh, and Rodney Zemmel, “Why products fail in Phase III,” In Vivo, April 2006.
Pharma R&D Compendium 2010 65
What gives drugs the X factor?

Despite the long development process, more than two-thirds of


What gives drugs the X factor? new drugs launched on the market are commercial failures. Recent
research uncovers what distinguishes those that succeed.

Ramesh Hariharan and Navjot Singh

Few industries have a more complex To understand better the causes of


value chain than the pharmaceutical commercial failure, we analyzed 50 new
sector. From the first stage of discovery molecular entities launched between 1997
to commercial launch, the R&D process and 2000. We considered six possible
is long (from 11 to 14 years per drug, factors that might influence their success,
on average) and expensive (from half and found that only two are important:
a billion to a billion dollars per drug). differentiation relative to the existing
To cover the costs, pharmaceutical standard of care, and market size.3 Of the
manufacturers traditionally rely on a few two, differentiation is the more significant.
blockbuster compounds to make up for
the underperformers. The findings have important implications
for a wide range of R&D, commercial, and
But the blockbusters are few and far business-development activities, both
between. Research has shown that only for compounds in development and for
about 30 percent of compounds entering those being launched. Post-launch failures
the market recover their risk-adjusted are bound to occur sometimes. But by
cost of R&D.1 understanding what drives commercial
success and so directing resources more
Why do so many branded drugs fail effectively, the industry as a whole—and
commercially? And what distinguishes certainly individual companies—can, we
the few that succeed? Companies have believe, achieve commercial success more
various theories. Some will point to the level than 50 percent of the time.
of unmet needs being addressed, others
to competition from branded drugs and Understanding
generics. Many believe that the novelty commercial success
of a compound’s mechanism of action
(MOA) is the critical factor. Yet few in the Pharmaceutical executives need a better
industry have studied the question explicitly. understanding of the variables that
Although the 70 percent failure rate at this determine the success or failure of product
stage is comparable to that of Phase II launches. We analyzed six potential drivers
clinical trials, post-launch failures receive of success.
far less attention. A 2004 study showed
that the novelty of a compound does not 1. Differentiation with respect to the
correlate with success in the market, but it standard of care in three areas: safety,
did not go on to examine other factors.2 efficacy, and convenience
66 Pharma R&D Compendium 2010 67
What gives drugs the X factor?

2. Novelty of the drug’s MOA, that is, a All the successful drugs reached annual during the flu season. However, it failed Why were the other variables insignificant?
binary assessment that depends on sales of $550 million, and most topped to meet the standard for convenience. We can offer rationales for each.
whether a commercial product with the $1 billion. Even in small markets, ten of Two chemicals, the active ingredient • The number of competitors is less
same MOA exists at the time of launch the 14 differentiated compounds were and lactose, needed to be placed in an important than how high a bar is set by
3. Market size at time of launch, measured successful; the only exceptions were four inhalation device before intake. the standard of care, and hence what
as the sum of all sales of drugs for a compounds that, despite being targeted at the company must do to exceed it.
given indication, along with an estimate a large market, ended up with narrow labels Additionally, we found that new entrants • We found no correlation between
of future growth4 restricting them to niche segments. need to exceed the standard of care only generic market share and success
4. Competitive intensity at time of slightly to achieve success. Products rates of new entrants; differentiated
launch, including the number of All 11 products that lagged behind the that are highly superior to incumbent products succeeded even in markets
patented competitors, the number of standard of care in any way were failures, compounds in safety, efficacy, or with strong generic share, reflecting
competitors in the pipeline, and the regardless of the market size. One drug convenience do not have a proportionately payors’ willingness to pay a premium for
market share of generics was approved and launched for the higher chance of commercial success. differentiation.
5. Unmet medical or market need, treatment of serious bacterial infections • Unmet needs that are not addressed by
assessed in the same three areas as and exhibited life-saving potential for An examination of the undifferentiated the new drug are irrelevant; the needs
differentiation: safety, efficacy, and some patients who did not respond products yields a surprise. All successful that are addressed are (at least partially)
convenience to the standard of care at the time of drugs in our data set of undifferentiated captured under differentiation.
6. Period of exclusivity, or the amount of launch. Yet the compound failed because drugs serve large primary-care markets. • Although success rates were greater
time until generic competitors can enter of a poor safety profile (severe cases Why? We believe one reason is that in markets with higher growth, the
the market. of inflammation and pain). Similarly, a specialist physicians tend to make more difference was statistically insignificant,
hyperlipidemia drug failed because it discerning recommendations than their probably because good brands can take
We analyzed these variables for 50 displayed lower efficacy than Lipitor, even primary-care counterparts. Those drugs market share from competitors even in
compounds released between 1997 and though it was safer by some measures that simply match the standard can stagnant markets.
2000 (see sidebar, “Methodology”). We (including lower risk of LFT elevation). succeed, therefore, provided they address • While patent duration will correlate to
then performed a regression analysis to Another product was launched with the large, primary-care markets and are lifetime profitability, it does not affect
identify the most important ones. goal of creating a market for a drug that marketed carefully. In addition, primary- the success of a launch, which is
expedited flu recovery. The drug was care markets tend to be large enough determined in the first few years.
Two variables—differentiation and market effective—the average recovery period fell to allow for well designed promotional
size—predicted the success or failure of all from five days to three—and even offered campaigns to exploit fine distinctions We found no correlation between
the compounds examined. Differentiation potential for off-label use for flu prevention between different segments of the market. differentiation and MOA. We expected
alone predicted the fate to find that compounds
of 77 percent of the EXHIBIT 1 EXHIBIT 2 with a novel MOA would
compounds, while market The power of differentiation and market size Novelty value be more likely to be
Novel mechanism of action is uncorrelated with differentiation
size accounted for the differentiated relative to the
remaining 23 percent. 14 drugs
▪ 10 (72%) were successful
10 drugs
▪ 10 (100%) were successful
12 drugs
▪ 10 (83%) were successful
12 drugs
▪ 10 (83%) were successful
standard of care. However,
The other variables were ▪ All 4 unsuccessful drugs ended
up with narrow labels in niche All differentiated products in large
▪ 2 (17%) were unsuccessful ▪ 2 (17%) were unsuccessful
among the 25 drugs with
markets (initially targeted markets were successful
statistically insignificant. High broader indications) High a novel MOA, only 12 are
Among the 25
novel drugs
differentiated—the same
As shown in Exhibit 1, all Differen- Differen-
almost an equal
number fall in
number of differentiated
ten of the drugs judged to tiation1 15 drugs
▪ 0 were successful
11 drugs
▪ 4 (36%) were successful
tiation1 13 drugs
▪ 6 (45%) were successful
13 drugs
▪ 0 were successful
the differentiated
and drugs as there are within
undifferentiated
be highly differentiated that – 7 inferior to standard of care
– 8 on par with standard of care
– All 4 were on par with standard
of care, and all in primary
▪ 7 (55%) were unsuccessful
categories the group of 25 with a
markets such as hypertension
were launched in moderate Low All undifferentiated and ▪ 7 (68%) were unsuccessful Low proven MOA (Exhibit 2).
inferior drugs failed in small – 4 inferior to standard of care (all
to large markets were markets failures)
– 3 on par with standard of care
More surprisingly, a larger
successful—that is, they percentage of drugs turned
were among the 25 with the Low Moderate to high Existing Novel out to be commercially
highest net present value of Market size2 Mechanism of action successful in the group
1
Average score on differentiation in efficacy, safety, and convenience. High is >3, low = 3, very low is <3.
the compounds we studied. 2
Market size at time of launch. Low is <$3 billion, high is >$3 billion. 1
Average score on differentiation in efficacy, safety, and convenience. Low is <3, high is >3. with the proven MOA than
Source: Interviews with experts; EvaluatePharma; analysts’ reports; McKinsey analysis Source: Interviews with experts; EvaluatePharma; analysts’ reports; McKinsey analysis
68 Pharma R&D Compendium 2010 69
What gives drugs the X factor?

in the group with a novel MOA (60 percent candidates and acquisition opportunities, Methodology
versus 40 percent). business-development teams often spend
a lot of time discussing variables that have We began our research with all 137 compounds launched from 1997 to 2000. We filtered this list to
Misplaced energy little influence on commercial success. exclude orphan compounds, non-NCEs (for example, reformulations), and drugs developed by small
manufacturers, leaving a sample of 94. Using an NPV analysis, we then identified the 25 top performers
Limited understanding of what drives Commercial resources dedicated at the (all of which had sales of at least $550 million; 23 had sales of more than $1 billion) and the 25 bottom
commercial success means energy and time of launch, and the expectations performers (all had sales of less than $200 million).5 For those 50 compounds, we rated five potential
resources are misallocated. For example, communicated to shareholders, are drivers of commercial success on a one-to-five scale, from “substantially inferior” to “substantially
instead of focusing on defining the often poorly aligned with the chance of superior.” The ratings were based on two or three expert interviews for each drug and external research.
current and emerging standard of care, commercial success. Even compounds To standardize the quantitative responses from experts, we were explicit in our definitions of each driver.
and on outlining a path to differentiation that are known to be inferior to the
In most cases, the expert evaluations were consistent with external articles. In the few cases where there
for clinical teams, much effort often goes standard of care (on any one dimension—
were disagreements, we supplemented the assessment with additional interviews. Our conclusions are
into sales forecasting—even though safety, efficacy, or convenience) get
based on the clinical judgement of these experts. We then conducted a regression analysis to assess the
the correlation between predicted and launched as long as they are approved.
relative importance of the potential drivers. We first chose the single variable that helped explain the
actual sales is poor—and the novelty of a Sometimes this is understandable given
compound’s MOA. sunk costs: it can pay to launch drugs largest number of successes and failures, then added variables sequentially to show the greatest possible
that are likely to generate positive returns incremental gain in prediction. We stopped adding variables when a sufficient number of compounds
When developing target product profiles from that point forward. But failing to were correctly described.
(TPPs), companies often rely strictly on deliver on expectations communicated at
We used logistical regression instead of linear regression, mainly because the former does not permit
detailed market research, which frequently the time of launch is not received well by
probabilities to be less than zero or greater than one. The resulting equation from the regression analysis
exaggerates the level that the new financial markets.
predicts a probability of success, which we then translated to “success” and “failure” depending on the
product must achieve to be successful.
In addition, some companies invest time Moreover, an uncertain understanding range of value (Exhibit 3). The analysis also demonstrates the law of diminishing returns: that highly
in developing a lengthy list of variables for of the value of differentiation causes differentiated compounds do not have a substantially higher probability of success than those that are
their TPPs, even though many of these are some companies routinely to put off life- only marginally differentiated relative to the existing standard of care (Exhibit 4).
unimportant to physicians. cycle management projects in favor of
developing new chemical entities.
Finally, limited understanding can result sometimes fail to understand adequately of the enterprise risk posed by some
in poorly designed clinical trials. For Finally, where companies do not the risk to future cash flows—the critical, late-stage assets is an important
example, development teams often do apprehend the commercial risk, they also enterprise risk. A solid understanding factor in capital structure decisions, such
not incorporate external as the optimal debt-to-
comparators (especially EXHIBIT 3 EXHIBIT 4 ILLUSTRATIVE equity ratio to maximize
the drugs that are the Regression model Diminishing returns shareholder value.
Probability of commercial success predicted by regression model Probability of commercial success (P)1
standard of care) in Phase
II and III trials, fearing Failures Uncertain zone Successful compounds
1.0
Further differentiation only slightly Implications
increases probability of success
that their compounds
0 0.2 0.4 0.6 0.8 1.0
will not stand up well to Aggrastat 0 Welchol 0.28 Flomax 0.67 Sustiva 1.00
Understanding what drives
them. While this might Tasmar
Relenza
0 Trovan
0 Agenerase
0.28
0.31
Actonel
Avelox
0.67 Zyvox
0.67 Xeloda
1.00
1.00 0.5 commercial success
Alamast 0 Zonegran 0.31 Femara 1.00
be wise in some cases, Tikosyn 0 Evoxac 0.33 Seroquel 1.00 suggests a clear set of
Rescriptor 0 Curosurf 0.33 Evista 1.00 Further differentiation significantly
some drugs destined for Synercid
Normiflo
0 Skelid
0.03 Hextend
0.33
0.33
Singulair
Kaletra
0.99
0.99
increases probability of success actions for executives.
Sonata 0.03 Actos 0.99
small, specialized markets Lotronex 0.03 Avandia 0.99
0
Corlopam 0.03 Mobic 0.99 1 2 3 4 5
would benefit from the Argatroban
Vitravene
0.03
0.03
Viagra
Celebrex
0.97
0.97 Substantially Marginally Undifferentiated Marginally Substantially Companies should
inferior inferior superior superior
comparison in clinical trials. Starlix
Rescula
0.09
0.10
Tamiflu
Temodar
0.90
0.90 emphasize clinical
Gabitril 0.10 Plavix 0.90 Differentiation
Fareston 0.10 Trileptal
Aciphex
0.89
0.87
differentiation rather than
Regression model and result
Similar problems crop up Protonix
Atacand
0.87
0.87
In [P/(1 – P)] = C0+ C1 average differentiation + C2 average unmet need + C3 market size + C4 mechanistic differentiation.
Avapro 0.87 competitive intensity + C5 generic share + C6 growth rate = -24.6 + 6.8 (average differentiation) +
in commercial activities. Micardis 0.87 1.1 (market size). This requires simple tools
1
Exact shape of the curve depends on the market size as per the above regression result. Each variable (for example,
In evaluating in-licensing differentiation, unmet need, market size) was normalized to a 1-5 scale based on physician interviews and/or market data. that can help summarize
Source: Regression model developed from ratings for compounds based on physician interviews; McKinsey analysis Source: McKinsey analysis
70 Pharma R&D Compendium 2010 71
What gives drugs the X factor?

the key differentiating attributes, and so development could focus more on


drive the debate around differentiation. understanding differentiation relative to
Understanding differentiation should take the standard of care at the time of launch,
priority over forecasting, managed-care and on the market size. (A longer list of
strategy, pricing, a detailed launch plan, attributes is more commonly the focus of
and life-cycle strategy, especially prior to extensive discussion.)
proof of concept.
Our research should help teams
In addition, for compounds for which spend their time and effort on the
efficacy results are available (typically after most important activities at the
Phase IIa), an appropriate commercial risk most appropriate junctures during
factor should be added to the technical development and product launch. It
risk of failure to increase transparency should thus also help to improve the
into the impact on future cash flows. quality of assessment, the quality of
the compounds being launched, and,
In early screenings of several possible therefore, the chance of commercial
in-licensing candidates, business success.

Ramesh Hariharan is an alumnus of McKinsey’s New Jersey office, and Navjot Singh
(navjot_singh@mckinsey.com) is a principal in the New York office.

We are grateful for the contributions made by Edd Fleming, Mike Pearson, Rodney Zemmel,
Michele Holcomb, Laura Furstenthal, Maria Gordian, Roy Berggren, Lara Sullivan, and Philip Ma,
all McKinsey colleagues. Padma Sundar deserves a special mention for conducting several
of the interviews and the initial analysis.

1
See Henry Grabowski, John Vernon, and Joseph DiMiasi, “Returns on R&D for 1990s new drug
introductions,” PharmacoEconomics, 2002 Vol. 20, pp.11-29.
2
See Bruce L. Booth Jr. and Rodney W. Zemmel, “The search for blockbuster drugs,”
mckinseyquarterly.com, August 2004.
3
Because our work is based on historical evidence, it assumes the status quo in development,
pricing, and payor environments. A radical shift in any of these areas could alter the primary drivers
of success for branded pharmaceuticals. The views in this paper are ours alone and are based
strictly on market data and interviews with physicians.
4
We measured market size and growth strictly by sales of all drugs for the indication in question,
providing us with an objective measure. We avoided using other possible measures of market size
(prevalence, number of patients being treated) as these are harder to assess and are only partial
components of a patient flow.
5
Costs of marketing and sales and R&D used to estimate the NPV are identical to those used by
Grabowski et al in “Returns on R&D for 1990s new drug introductions,” PharmacoEconomics,
2002 Vol. 20, pp.11-29.
Pharma R&D Compendium 2010 73
A special kind of success

Mainstream pharmaceutical companies are turning their


A special kind of success attention to specialty drugs. Which factors are most
important for commercial success or failure?

Mark Gudiksen, Edd Fleming, Laura Furstenthal, and Philip Ma

For most of the past 30 years, large advancing from 39 percent of total sales in
pharmaceutical companies have focused 2001 to 45 percent in 2006 (Exhibit 1).
mainly on developing and marketing
blockbuster drugs that are used by Although specialty products traditionally
extensive populations of patients. These have been the preserve of biotechnology
drugs, such as Lipitor (atorvastatin, Pfizer), pioneers such as Genentech and Amgen,
Nexium (esomeprazole, AstraZeneca), mainstream pharmaceutical companies
and Zoloft (sertraline, Pfizer), are usually are increasingly adjusting their focus. The
prescribed by primary-care physicians chief executive officers of Pfizer, Bristol-
(PCPs). During the past decade, Myers Squibb, and AstraZeneca have
however, the industry has seen the all publicly announced their intention to
growing commercial success of specialty increase their development of specialty
pharmaceuticals—drugs prescribed mainly and biologic products.
by specialists rather than PCPs. Total
annual revenue in the period from 2001 to Given the excitement surrounding specialty
2006 of all drugs launched between 1994 drugs, we set out to investigate the
and 2003 shows that specialty products are common factors that contribute to their
taking an increasing portion of the market, success. For example, technological
innovations—such as
EXHIBIT 1 recombinant proteins,
Specialty drugs’ share of the market monoclonal antibodies,
Total sales from pharmaceuticals launched 1994–2003, Specialty
$ billion Primary care and genome-based
Annual
diagnostics—tend to make
Market value
growth rate, % their commercial debuts
260
240
243 26 in specialty markets,
220 216 indicating a greater focus
200
180
183 45 30 on novelty in product
160
144
45
development. At the
44
140
same time, modifications
120
42
100
103
of existing drugs, such
77 40
80
39 55 23 as new formulations,
60 55
40
60
58
56
are quite common in
61
20
specialty markets given
0
2001 2002 2003 2004 2005 2006 manufacturers’ desire
to preserve franchises
Source: EvaluatePharma; McKinsey analysis
74 Pharma R&D Compendium 2010 75
A special kind of success

through lifecycle management and the drawn from various commercial databases The drug rating system
wish of specialty physicians for improved (Pharmaprojects, EvaluatePharma,
To find out which factors were important in deciding the success and failure of the drugs in our study, we
therapeutic options for their patients. So and IMS). We then decided which
rated each product according to various criteria and then used regression analysis. To give equal weight to
which factors are the most important for of these parameters correlated
all factors for the regression analysis, we rated each product using a five-point scale.
success or failure in this field? with commercial success using the
technique of logistic regression. Differentiation versus standard of care (in terms of efficacy, safety, and convenience) at launch. 1,
Basis of analysis substantially inferior; 2, slightly inferior; 3, at parity; 4, slightly superior; 5, substantially superior. Source:
Differentiation is the key interviews with physicians
We analyzed all 143 non-generic, specialty
pharmaceuticals launched in the United Our analysis shows that only three of the Market size at launch of primary indication. 1, <$0.25 billion; 2, $0.25-1.5 billion; 3, $1.5-3 billion; 4, $3-5
States from 1994 to 2003, using the net above factors were required to explain billion; 5, >$5 billion. Source: EvaluatePharma; McKinsey analysis
present value (NPV) of the first five years the commercial success of 96 percent
Market’s annual growth rate over five years post-launch. 1, <0 percent; 2, 0-10 percent; 3, 10-20 percent; 4,
of US sales post-launch as our metric of (69 out of 72) of the specialty drugs we
20-30 percent; 5, >30 percent. Source: EvaluatePharma
commercial success. We classified drugs examined (Exhibit 2).
as specialty pharmaceuticals or primary- Competitive intensity (for example, number of competing products available at launch). 1, >6; 2, 5-6; 3, 3-4;
care products by interviewing physicians to The most important is differentiation relative 4, 1-2; 5, 0. Source: EvaluatePharma; The Medical Letter; interviews with physicians; McKinsey analysis
find out whether specialists or PCPs were to the standard of care in terms of efficacy,
the primary prescribers. Taking the most safety, and convenience. This accounts for Novel mechanism of action (MoA). 1, simple reformulation; 2, complex reformulation (for example,
successful quartile (36 drugs, five-year NPV 51 percent of a specialty drug’s success. pegylation); 3, novel MoA, five to ten years after lead; 4, novel MoA, two to five years after lead; 5, novel
of US sales ≥ $900 million, median fifth- In particular, the top 36 products were all MoA, lead candidates. Source: EvaluatePharma; Pharmaprojects; interviews with physicians; The Medical
year US sales ~ $590 million) and the least significant improvements on the treatments Letter; McKinsey analysis
successful (36 drugs, five-year NPV of US available at the time of launch. Successful
sales < $140 million, median fifth-year US products did not have to improve on all Disease severity/unmet need. 1, no risk of morbidity; 3, some severity; 5, high morbidity. Source: interviews
with physicians
sales ~ $20 million), we assigned scores to aspects of efficacy, safety, and convenience;
each drug according to various measures many were superior in one respect only and
(see sidebar, “The drug rating system”). close to parity for the others. to have a successful product in a large, in our survey that had small markets at
• Differentiation versus standard of existing market than to create a market for launch (≤$250 million in annual sales) and
care at time of launch in terms of Market size at launch is the second most a new product—which helps to explain big found that the successful ones were highly
efficacy, safety, and convenience important factor, accounting for 25 percent pharma’s focus on “me-too” products. This differentiated (average differentiation of
• Market size at launch of success. Simply stated, it is easier finding is supported by the work of Ramesh 4.0 on a 5-point scale) with respect to the
• Five-year market Hariharan and Navjot Singh, which showed standard of care at launch and were focused
growth rate EXHIBIT 2 that market size at launch and differentiation on severe diseases with high unmet need
• Number of competing Success factors for specialty pharmaceuticals were the only factors correlating with the (Exhibit 3). By contrast, products that were
Success or failure predicted by adding variable,
drugs at launch % success of drugs made by large companies. unable to differentiate themselves sufficiently
• Degree of novelty 100
from the pre-existing standard of care, with
4
in the mechanism 90
19
Market growth rate over the first five an average differentiation slightly lower than
of action (MoA) years after launch is the third vital factor the standard of care (average differentiation
80

70

• Disease severity 60
51
25
for success with 19 percent correlation, of 2.9), were failures.
and unmet need. 50

40
showing that specialty products have the
30 capacity to be “market creators.” Rituxan In addition to understanding which factors
We assigned these 20

10
(retuximab), Genentech’s revolutionary influence success, it is equally important
scores on the basis of 0 treatment for non-Hodgkin’s lymphoma, to understand those that do not. Although
Average Market size at Five-year Other1
interviews conducted differentiation
relative to
launch market growth
rate
is an example of a drug that was able to specialty products are often considered
standard of care
with about 50 expert 1
expand a market from about $20 million in highly novel, novelty did not correlate
The model did not predict accurately the success or failure of three products. Gemzar (gemcitabine, Eli Lilly) was predicted as
physicians, consultation a success, but this rests on its status as the only treatment available for pancreatic cancer; physicians are looking for a better
product. Aldurazyme (laronidase, BioMarin/Genzyme) was predicted as a failure but is an effective treatment for Hurler’s
total sales to $1.5 billion in only five years. with success. This indicates that a novel
of the primary medical disease (mucopolysaccharidosis type 1), although it has a very small market. Femara (letrozole, Novartis) was predicted as a
failure but is successful when used as adjuvant therapy with tamoxifen, as shown by post-marketing clinical trials released in
To understand this phenomenon of market MoA does not necessarily amount to
literature, and information 2003. However, as Femara was launched in 1997 it rates as unsuccessful according to the criteria of this study, which
focuses on the first five years after launch. creation better, we analyzed all the drugs differentiation from a physician’s perspective.
Source: McKinsey analysis
76 Pharma R&D Compendium 2010 77
A special kind of success

EXHIBIT 3 looked at the correlation of commercial widely used HIV drugs) are the three
Specialty drugs launched into small markets success with regard to efficacy, safety, therapeutic areas that dominate the
Drugs launched 1994-2003 with initial market sizes ≤$250 million 1

Average
Market size
at launch,
Market
five-year
Novelty of
mechanism Disease and convenience. Exhibit 4 shows that landscape of specialty pharmaceuticals,
Drug Company2 differentiation $m CAGR,3 % of action severity
Rituxan (rituximab) Genentech/Biogen Idec 4.2 23 132 5 5
novel molecules are more likely to be representing more than 65 percent of
successful if they demonstrate improved all specialty drugs in our survey (Exhibit
Successful drugs

Synagis (palivizumab) MedImmune 4.6 142 46 5 5


Aricept (donepezil) Eisai 4.2 236 37 1 4
Gleevec/Glivec (imatinib)
Camptosar (irinotecan)
Novartis
Pharmacia
4.8
3.4
257
72
68
74
5
5
5
5 efficacy, follow-on drugs if they show 5). However, their respective rates
ReoPro (abciximab) Eli Lilly 3.5 23 86 5 4
Gemzar (gemcitabine) Eli Lilly 3.2 62 64 1 5 improved safety, and reformulations if of success and failure are drastically
Average 4.0 116 72 3.9 4.7 they offer enhanced convenience. The different. Oncology has significantly more
Cervidil CR (dinoprostone) Forest Laboratories 3.3 18 12 1 1
result for follow-on products is particularly (42 percent) of the failures, but only 28
Aldurazyme (laronidase) BioMarin/Genzyme 4.2 12 63 5 5
ProAmatine (midodrine) Shire 2.5 230 -2 5 5 striking, as we found not only that an percent of the successes, with success
Trisenox (arsenic trioxide) Cephalon 3.2 138 23 5 5
Infasurf (calfactant) Forest Laboratories 3.2 154 0 1 5
improved safety profile accounts for 34 most strongly tied to improvements in
Ontak/Onzar (denileukin diftitox) Eisai 3.3 28 12 5 5
percent of their success (improvements efficacy. The large number of oncology
Refludan (lepirudin) Schering AG 2.8 20 46 3 4
in efficacy being generally irrelevant, failures may be due to the rapidly
Unsuccessful drugs

Lustra/Lustra-AF (hydroquinone USP 4%) Medicis Pharmaceuticals 2.7 11 6 1 1


Kinerase (kinetin) Valeant 3.1 31 58 5 1
Vistide (cidofovir) Gilead 2.4 221 -4 3 5
with a 3 percent correlation rate), but improving standard of care in oncology,
Salagen (pilocarpine) MGI Pharma 2.7 4 39 1 1 also that 77 percent of the products driven by the extreme heterogeneity
Cetrotide (cetrorelix) Serono 2.8 11 23 5 1
Carticel (autologous cultured chondrocytes) Genzyme 2.2 254 11 5 4 were successful compared with about of cancer and the large numbers of
Gliadel Wafer (polifeprosan 20 with carmustine implant) Guilford Pharmaceuticals 1.9 45 52 2 5
Oncaspar (pegaspargase) Enzon 3.4 3 11 2 5
40 percent of novel products and associated pathways and targets, which
Photofrin/Photobarr (porfimer sodium) Axcan Pharma 3.0 87 57 5 2 reformulations. One implication is that, leads to market fragmentation. For
Ethyol (amifostine) MedImmune 2.4 22 89 5 2
DepoCyt (cytarabine liposome injection) Enzon Pharmaceuticals 3.5 4 12 1 5 in certain markets (see discussion below instance, many of the failures ended
Average 2.9 93 13 4.1 4.0
on therapeutic areas), taking time to up as treatments for limited patient
refine the safety profile and aiming for populations: Ontak/Onzar (denileukin
1
Some specialty drugs were able to create markets through significant differentiation and focus on severe disease areas.
2
Company or companies that owned the product during first five years after launch.
3
Compound annual growth rate.
Source: EvaluatePharma; Pharmaprojects; The Medical Letter; physician interviews; McKinsey analysis more desirable indications may be a diftitox, Ligand Pharmaceuticals/Eisai),
better strategy than rushing to be first. Trisenox (arsenic trioxide, Cephalon),
For instance, inspection of the market- We segmented all of the products into and Oncaspar (pegaspargase,
creating products (Exhibit 3) shows that three groups based on their novelty: first, Not all therapeutic Enzon) were all indicated for very low-
successful products did not, on average, lead-candidate molecules with novel areas are equal incidence leukaemias or lymphomas.
have more novel mechanisms than the MoAs; second, follow-on products,
failures. The number of competing products launched two to ten years after the Oncology (including immunomodulators), Also, although oncology has been a hotbed
did not affect success either, illustrating lead molecules; and third, precedented the central nervous system (CNS), and of innovation—Rituxan, Gleevec (imatinib,
again that it is only a product’s relative mechanisms and reformulations. We then systemic anti-infectives (in particular, Novartis) and Herceptin (trastuzumab,
merits that are significant Genentech)—some of the
(that is, its differentiation EXHIBIT 4 EXHIBIT 5 most successful products
relative to the standard of Correlation between degree of novelty and key success factor Dominant therapeutic areas for specialty pharmaceuticals were based on well-known
Breakdown of success and failure by therapeutic area
care) and not the number Number of drugs, % mechanisms with indication
Number Drugs, Efficacy, safety, and convenience,
of competitors faced. Novelty of drugs % of successful drugs in category % correlation of success 1
expansions. These include
72 36 36 = 100%
38 100 143 Eloxatin (oxaliplatin, Sanofi
First place does A. Novel mechanism
of action, lead 29 44
19
10
90
28
Oncology and
immunomodulators
Aventis), the first platinum
not always win candidates 35
(29 drugs)
80 31 42 compound to show
34
70
significant effects against
60 Central nervous
25
Although our findings B. Follow-on, 2–10
14 18
11
system colorectal cancer, and
50
indicate that novelty years after lead 13 77
3 3 40
14 6 Gemzar (gemcitabine, Eli
14 3 Systemic
itself is not an indicator 30
8
7
6 22
anti-infectives Lilly), the first pyrimidine
5 11
of success, we did find 34
20 5 7
11 Blood
analogue with effects
C. Precedented 6
that it might provide mechanisms of
action and
30 43 10
8 10 19 22 8
Musculoskeletal
Dermatology
on pancreatic cancer,
14 0
a way for companies Other
showing that novelty is
1
reformulations 6
0
Efficacy Safety Convenience
All drugs Top failures Failure Success
to understand how to and successes not a requirement for
position their products. success in oncology.
1
Correlation is the R2 of the linear regression of the net present value of the first five years of sales with
efficacy, safety, and convenience. 1
Other includes cardiovascular, endocrine, genito-urinary, respiratory, gastrointestinal, and metabolic.
Source: EvaluatePharma; Pharmaprojects; The Medical Letter; physician interviews; McKinsey analysis Source: EvaluatePharma; McKinsey analysis
78 Pharma R&D Compendium 2010 79
A special kind of success

In the field of CNS drugs, specialty associated with identifying new CNS delivery can produce results. We suggest An additional question is whether or
products have fared much better, targets, our research highlights that the that several possibilities exist to increase not these findings will continue to ring
representing 25 percent of successes and CNS is a therapeutic area in which the the likelihood of success in the specialty true considering the many potential
only 11 percent of failures. The former are rewards and success rates may warrant market. First, include more practising changes in the pharmaceutical
mainly antipsychotics (55 percent), with the more significant exposure. clinicians in the process of defining the landscape in the next few years. With
rest being treatments for multiple sclerosis, target product profile to bring about the rise in personalized medicine,
epilepsy, and Alzheimer’s disease. Anti-infectives/HIV therapies have a similar a more practical assessment of the increased pressure from payors for
Because many CNS-related diseases profile to CNS drugs in that they represent true value of a new drug over the likely “pay-for-performance” therapies,
are chronic conditions, safety played a more successes (22 percent) than failures standard of care at time of launch. Second, and the probable greater use of
major role in deciding which products (6 percent). As HIV has been transformed encourage early-stage development pharmacoeconomic analysis, the hurdles
succeeded. In particular, none of the five into a chronic disease, so successful teams to articulate potential approaches for new drugs are likely to multiply
successful antipsychotics in our survey— therapies have come to be distinguished to differentiation during portfolio reviews. rather than diminish. If this proves to
Zyprexa (olanzapine, Eli Lilly), Risperdal from earlier drugs by an enhanced safety Third, ensure the right trade-offs are made be the case, then the need to create
(risperidone, Janssen), Abilify (aripiprazole, profile. Reyataz (atazanavir, Bristol-Myers (especially between safety and efficacy) in differentiated products—and knowing
Otsuka/Bristol-Myers Squibb), Seroquel Squibb) and Viread (tenofovir disoproxil designing clinical trials, particularly for drugs how to surpass the future standard of
(quetiapine, AstraZeneca), and Geodon fumarate, Gilead) are successful largely that are not first to market. care—will be more important than ever.
(ziprasidone, Pfizer)—was viewed by because they have fewer side effects than
physicians as having any real efficacy some of the older, comparable therapies. A version of this article, entitled “What drives success for specialty pharmaceuticals?” was first
advantage compared with others (all had In addition to the safety profile, several HIV printed in Nature Reviews Drug Discovery, Volume 7, pp. 563–567 (June 6, 2008). For further details
average efficacy scores of 3.0), but all drugs—Combivir (lamivudine + zidovudine, of the methodology and analysis, please contact the authors.
were perceived as having various safety GlaxoSmithKline), Trizivir (abacavir +
benefits. zidovudine + lamivudine, GlaxoSmithKline), Edd Fleming (edd_fleming@mckinsey.com) and Philip Ma (philip_ma@mckinsey.com) are directors
and Kaletra (lopinavir + ritonavir, Abbott) in McKinsey’s Silicon Valley office, where Laura Furstenthal (laura_furstenthal@mckinsey.com) is a
Another interesting CNS success story were successful owing to the convenience principal. Mark Gudiksen is an alumnus of the San Francisco office.
is Aricept (donepezil, Eisai/Pfizer), benefits of combination therapies, which
which has become the standard of reduced the number of pills taken by the The authors wish to thank Jay Chiang, Jerel Davis, Padma Sundar, and Denise Flaherty for their
care for Alzheimer’s disease. Its efficacy patient. Emtriva (emtricitabine, Gilead) assistance in preparing this article.
was viewed by many physicians as provides an interesting example of the
“unremarkable,” but, owing to a good benefits of the combination pill to the
safety profile and a lack of other treatment patient; formulated in isolation it is one
options, they also commented that they of the “failures” on the list because of
did not see “any harm” in prescribing it. its “inconvenience.” However, it is also
a component, along with Viread, of the
In some regards, it is surprising to note highly successful combination pill Truvada
the relative success of CNS drugs, a (which launched in 2004, so is not
therapeutic area that many companies included in this analysis).
target less aggressively, owing to the
more limited understanding of the Winning strategies
underlying biology and hence the
narrower set of known “druggable” Our research suggests that what really
pathways and targets. For example, matters to specialty physicians is creating
the antipsychotics all target some a therapy that is a significant improvement
combination of serotonin and dopamine over the standard of care. Novel MoAs
receptors, whereas the antiepileptics might represent exciting science but are
mainly target various pathways involving not correlated with differentiation from the
the GABA (γ-aminobutyric acid) receptor. standard of care, whereas less “exciting”
Nevertheless, even given the challenges approaches such as reformulating drug
Pharma R&D Compendium 2010 81
R&D in emerging markets: a new approach for a new era

Gone is the time when R&D in developed markets could meet the
R&D in emerging markets: pharmaceutical needs of emerging markets. Local presence is required.
a new approach for a new era
Michael Edwards

The pharmaceutical industry has long Instead, they should consider how R&D
been driven by the demands of the north can best serve these disparate and
American and west European markets, with often poorly understood regions. The
scant attention given to emerging markets likely outcome is that they will place a
and the diseases more prevalent there. significant proportion of their scientists,
their chemistry, manufacturing, and
This is changing. Emerging markets controls, and their development groups
contributed 30 percent of the there. Only in this way will they understand
pharmaceutical industry’s value in 20081 the needs and preferences of patients and
and are forecast to grow by 14 percent physicians, develop products that meet
a year to 2013.2,3 By contrast, the US those needs, and ensure the products are
market—which still represents 40 percent introduced in a timely manner.
of the global market—is expected to grow
more slowly with annual growth of less Medical needs and preferences
than 3 percent over the next four years.
Indeed, all developed markets tend to be Patients in emerging markets have
characterized by low or negative growth, a different medical needs from patients in
stricter regulatory environment, increasing the West, and they and their physicians
patent litigation, and greater scrutiny of have different treatment preferences. The
drug safety. result is demand for products that reflect
these variations. This affects decision
Not surprisingly, many pharmaceutical making and resource allocation across
companies now see some of the largest pharmaceutical companies’ entire R&D
emerging markets as the key to their value chain, from basic research to in-
growth ambitions. Over the past ten years, market investment. There are several areas
they have established low-cost research to consider:
institutions in emerging markets, tapped into
talent pools there in search of innovation, Choice of disease area
and set up corporate social responsibility The prevalence of some diseases varies
programs to help tackle neglected diseases. significantly between geographic regions
But more needs to be done if companies are and ethnic groups. This can be due to
to capture the opportunity. genetic differences or environmental
factors such as diet and living conditions.
Most still tend to regard emerging markets Diseases that mainly affect developing
mainly as a source of lower-cost R&D. countries and that are not prevalent in
82 Pharma R&D Compendium 2010 83
R&D in emerging markets: a new approach for a new era

developed markets have been largely markets, with the exception of TB in India.5 whereby the dose is tailored to genetic physician and the patient, and they
ignored from a pharmaceutical “for profit” Their value, while growing, will remain differences between individuals rather than can improve patients’ compliance.
viewpoint. However, as economies in small in the near term by developed groups, is still some way off. Thirty percent
emerging markets grow and governments market standards, though some resources of Asians, for example, have a variation in The polypill, manufactured by India’s
begin paying for treatment for the poor, are being applied to dealing with TB. their cytochrome P4502C19 gene which Cadila Pharma, includes five APIs and
certain hitherto neglected types of disease AstraZeneca, for example, has established results in reduced ability to metabolize up is one of the most extreme examples of
present revenue opportunities. an R&D center in Bangalore dedicated to to 15 percent of all clinically useful drugs,6 a fixed-dose combination. The polypill,
TB. Research into these diseases is often compared with 6 percent of Caucasians. which is in Phase III, will be targeted at
The better to understand these diseases, conducted in conjunction with public and This influences clinical practice. Physicians patients with elevated blood pressure
companies are realizing that they need charitable bodies, or in the context of in Hong Kong commonly prescribe or high cholesterol, and combines
dedicated R&D resources in emerging access-to-medicine programs. lower doses of certain drugs—diazepam hydrochlorthiazide, atenolol, ramipril, and
markets, as valuable insights from patients, (Valium) is one—for Chinese patients simvastatin, with low-dose aspirin and folic
physicians, and payors can be gained In the case of “diseases of poverty” such than for Caucasians, because individuals acid.8 The polypill therefore includes APIs
only through local presence. Currently, as the dehydrating illness cholera, it is carrying this variant are at higher risk of originally made by AstraZeneca, Sanofi-
oncology is the therapeutic area with the not so much research that is needed as experiencing toxicities when taking these Aventis, and Merck (although all the drugs
most significant commercial opportunities better basic living standards. For sufferers drugs. In addition, the efficacy of some are now available as generics). Already on
for drugs developed specifically for emerging of cholera, the treatment is as simple as drugs is limited to certain genetically the market is the CV Pill, made by Torrent,
markets. Hepatocellular carcinoma (HCC), having access to clean drinking water and defined populations or ethnicities, and as a which combines atorvastatin, ramipril,
often caused by hepatitis B and C virus rehydration powder. result regulators might approve a drug only metoprolol, and aspirin. In some markets
(HBV/HCV) infection, is the most frequently for particular ethnic groups. BiDil—a fixed- these combinations can be priced at a
cited example. HCC is the fourth most Efficacy and dosing dose combination of isosorbide dinitrate premium and win patent protection.
common cancer globally, and 75 percent Dosing levels may need to be adjusted and hydralazine used to treat heart
of the one million people affected each in emerging markets to reflect variations failure—was dismissed when it failed to Formulations represent another
year live in east Asia. Other cancers more in the efficacy and toxicity of a drug at a demonstrate efficacy in a heterogeneous opportunity to meet local needs while also
prevalent in developing countries include given dose between different geographical population trial, but revived when efficacy potentially extending a product’s lifecycle.
Kaposi’s sarcoma—less frequently seen in or ethnic populations. For example, some was shown in patients with significant There are many formulations that can be
developed markets since the advent and subpopulations or ethnic groups have African ancestry.7 considered, including extended-release,
use of the HIV “triple cocktail”—and gastric/ lower levels of critical P450 enzymes, soluble powders, inhalers, nasal sprays,
oesophageal cancers, which have much which affect drug metabolism and As with disease areas, real insight into the injectables, and long-acting patches. In
higher prevalence in east Asia. HBV infection therefore dosing requirements. nuances of dosing can be gained only each case, the benefits of meeting an
represents another opportunity: of the 400 through close interaction with local experts emerging market need or preference
million people infected with HBV worldwide, Body mass also affects dosing levels, and and local experience. have to be weighed against the cost
roughly one-third are in China. Many populations in emerging markets tend to of developing a novel formulation, and
pharmaceutical companies already focus have lower average body mass than those Patient and prescriber preferences thought given to whether the formulation
on some of these opportunities. Novartis, in the United States or Europe. In Japan, Understanding physicians’ and can be developed in time to extend the
for example, is investing in an R&D center local regulators increasingly approve patients’ preferences and responding lifecycle adequately.
in Shanghai, which will initially focus its lower doses than their counterparts in to them effectively is important to
research on diseases particularly common in the United States. Approved doses for success in emerging markets. Fixed- The logistics of delivering products to
China (including hepatitis B and C).4 Bayer’s Ciprofloxacin, for instance, vary by dose combination medicines—drug hospitals and pharmacies is another
region—600 mg in Japan, up to 800 mg in therapies with two or more active important area that pharmaceutical
Other neglected diseases include TB, the United Kingdom, and up to 1,200 mg pharmaceutical ingredients (APIs) companies will need to understand. For
malaria, roundworm, and Chagas. in the United States. combined into one tablet—are preferred example, heat and humidity can destroy
Although these diseases are a major in many markets, but their popularity medications, and there may not be a
health problem across large parts of the Genetic differences between certain still varies greatly by individual market. reliable cold-chain supply system to
world, they are not among the 20 most ethnic groups are a further factor in dosing Their benefits are that they can be deliver medication from factory to hospital.
prevalent diseases in the largest emerging levels—although personalized medicine, more convenient for the prescribing In 2007, in Uganda, Abbott launched
84 Pharma R&D Compendium 2010 85
R&D in emerging markets: a new approach for a new era

Aluvia, a heat-stable tablet formulation the total patient numbers required to and attentiveness to specific needs. In By building significant R&D resources
of the protease inhibitors lopinavir and satisfy US and European regulators. countries that do not require inclusion of in emerging markets, pharmaceutical
ritonavir. The soft-gel capsules and oral local patients as a condition of regulatory companies will revolutionize the
solution that were previously available By including relevant populations from approval, local Phase IV post-launch global R&D group, helping shift the
were not appropriate due to the scarcity emerging markets in trials, companies trials or epidemiological studies can help focus of the entire development
of refrigeration equipment. The tablet might also be able to reduce the current demonstrate efficacy in the immediate pathway—from early development to
version has now been filed for registration time delay that exists between launching patient population. (The number of Phase IV lifecycle management—to include both
in more than 150 countries, and nearly their drugs in the West and in emerging trials in emerging markets has grown at an developed and developing markets.
75,000 patients were treated in 2008. markets—a delay that can mean the annual rate of more than 50 percent in the
loss of significant patent revenue. Some past five years.) Post-launch activities, such For the pharmaceutical industry it will
Coming to market companies are beginning to tackle this as Phase IV trials in new indications, might mark the end of an era when it was
issue. Merck, for example, has made a also lead to expanded indications, even on assumed that the needs of the world’s
Regulatory requirements and the public commitment to launch its drugs a local basis, providing the sales force and pharmaceutical market were largely those
move towards global trials simultaneously in both spheres—no medical scientific liaison staff with additional of developed countries. The new era
Establishing R&D activities in emerging small undertaking given that some of its scientific data for their discussions with will require considerable organizational
countries will help pharmaceutical previous launches in emerging markets local doctors, and reinforcing the message change. R&D departments will need new
companies obtain market access have been up to ten years behind those in that the company is focused on meeting resources. And since emerging market
in a timely way. To a certain extent, the West. Other companies also appear to the needs of the regional market. revenues are likely to be less than half
some local presence is already a be moving towards this goal: Bayer was those of the United States and European
prerequisite. China, India, South Korea, selling the multi-targeted kinase inhibitor Local R&D activities also help build Union in the medium term, funding might
and Taiwan all require trials with local sorafenib (Nexavar) for HCC in China relationships with governments and have to be ring-fenced. But perhaps the
patients for local product registration. less than a year after its US launch, while regulatory agencies. In some places, a most important change of all, given the
Some pharmaceutical companies Bristol-Myers Squibb launched the antiviral local presence is already a prerequisite for speed at which the market is developing,
use contract research organizations HBV therapy entecavir (Baraclude) in market access, as is increasingly the case will be a rapid change in mindset.
to meet these requirements, but China with only a six-month delay. in Russia; elsewhere, it helps to establish Companies already recognize the need to
others have established their own relationships with the authorities that might tailor their product portfolios for emerging
development centers to run the Market shaping through local R&D accelerate approval of a drug and deliver markets, but to be real leaders in the future
trials. Sanofi-Aventis, for example, Conducting R&D in emerging economies higher reimbursement. they must tailor their R&D activities too.
established a Biometrics Research provides a number of market-access
Center in Beijing in 2008 to support benefits to pharmaceutical companies Michael Edwards (michael_edwards@mckinsey.com) is an associate principal in McKinsey’s London office.
global and local clinical trials, from the by engaging local stakeholders
initial design of studies through to data early in drug development.
management and statistical analysis.
Working with leading physicians at this
Local requirements apart, there are stage can help build a product’s name and,
other benefits of including emerging most importantly, give companies insights 1
IMS World Review, 2009.
markets in trials for new drugs. into how to tailor products to local needs. 2
IMS Health, Market Prognosis, March 2009.
Development cycles can be reduced For example, working with local physicians 3
“Global pharmaceuticals: emerging markets—infusing badly needed revenues for years to come,”
owing to faster recruitment of subjects in east Asia to develop new oncologic Bernstein Research, May 2009.
from a larger pool of patients. And the drugs (such as tyrosine kinase inhibitors) 4
See http://www.novartis.com/research/pharmaceutical.shtml.
costs of recruiting patients and paying could help pharmaceutical companies learn 5
“Mortality and burden of disease estimates for WHO member states in 2004,” World Health
investigators are lower. This can benefit how local oncologists approach cancers Organisation, February 2009.
both the development of drugs that such as GI stromal tumours, which are rare 6
“Racial differences in response to pharmacological treatment,” Chung, A, 2004.
are specifically targeted at emerging in developed countries. 7
Abdallah S. Daar and Peter A. Singer, “Pharmacogenetics and geographical ancestry: implications
markets and that of drugs for developed for drug development and global health,” Nature Reviews, 2005, Vol. 6, 2005.
markets, as patients recruited for trials in A local R&D presence also helps build 8
“Effects of a polypill (Polycap) on risk factors in middle-aged individuals without cardiovascular
emerging countries can count towards a company’s reputation for innovation disease (TIPS): a Phase II, double-blind, randomized trial,” The Indian Polycap Study, The Lancet,
2009, Vol 373.
Pharma R&D Compendium 2010 87
The microeconomics of personalized medicine

Personalized medicine promises to increase the quality of


The microeconomics of clinical care and, in some cases, decrease health care costs.
personalized medicine The biggest hurdles are economic, not scientific.

Jerel Davis, Laura Furstenthal, Amar Desai, Troy Norris,


Saumya Sutaria, Edd Fleming, and Philip Ma

Personalized medicine—defined here as Our investigation highlighted three


a tailored approach to patient treatment main obstacles to the advancement of
based on the molecular analysis of genes, personalized medicine: first, scientific
proteins, and metabolites—has generated challenges (poor understanding of
much excitement. Yet few personalized molecular mechanisms or a lack of
medicine tests have achieved high molecular markers associated with
clinical adoption. To understand better some diseases, for example); second,
the challenges to the development and economic challenges (that is, poorly
acceptance of personalized medicine, and aligned incentives); and third, operational
how to overcome them, we interviewed issues (electronic tracking of diagnostic
more than 60 leading payors, providers, information, privacy concerns,
regulatory experts, pharmaceutical and reimbursement coding problems, provider/
biotechnology companies, academic patient education). Although scientific
opinion leaders, and diagnostics and difficulties remain, it now seems that the
clinical laboratory companies, and economic challenges and operational
conducted microeconomic analyses of questions are the biggest hurdle. In many
different stakeholder issues (see sidebar, cases, operational issues can be largely
“Research details”). resolved within a particular stakeholder
group. However, correcting
EXHIBIT 1 the incentive structure and
Not all diagnostic tests save costs for payors1 modifying the relationships
between stakeholders
Procedure-

Companion diagnostics
focused
diagnostics Genetic risk markers
could be more complex.
KIF6
Her22 BCR-ABL3 Warfarin AlloMap BRCA1 BRCA1-F6 (statin)
Savings from
changed decision,
In this article, we discuss
40 80 2 4 25 25 -3.5
$ thousands
the economic challenges
×
Probability that
diagnostic changes 70 5 35 75 2 20 50 of personalized medicine
treatment decision, %
= from the perspective
Savings per test,
$ thousands 28 4 0.7 3.1 0.5 5 <0 of four key groups of
stakeholders: payors,
Cost of test,
$ thousands 0.1 1 0.3 3 2–3 2–3 na
providers, pharmaceutical
Cost saving for
and biotechnology
payors? 9 9 9 9 8 9 8 companies, and
1
Estimated savings per test is the product of savings from a single changed treatment decision and the probability that any diagnostics companies. We
given patient will have a “positive” test (such that treatment decision is changed).
2
3
Human epidermal growth factor receptor 2. focus on the US market
Breakpoint cluster region–abelson tyrosine kinase.
88 Pharma R&D Compendium 2010 89
The microeconomics of personalized medicine

in particular, but the matters raised are Two primary factors determine a test’s cost basis. Variants in KIF6, for example, have interests to delay adopting personalized
also relevant elsewhere. Our proposals effectiveness from a payor’s perspective: been linked to a 50 percent increase in the medicine tests until they can differentiate
for overcoming these challenges could per patient savings (that is, the difference risk of myocardial infarction, but this risk between those that are cost saving and
significantly accelerate the adoption of between the cost of treating the disease can be reduced to normal levels through others that are cost creating. The winning
personalized medicine. and the cost of the intervention indicated treatment with statins.1,2,3 Widespread strategy for diagnostics companies may
by the test); and the likelihood that a test use of a hypothetical test based on these therefore be to collaborate with other
Stakeholder incentives suggests an intervention for any particular markers could actually result in higher stakeholders where the economics
and challenges patient (Exhibit 1). Tests that help avoid the costs through treating patients with are more aligned (for example, Kaiser
use of expensive therapies (for example, statins, compared with the savings from Permanente, large self-insured employers,
Payors trastuzumab [Herceptin, Genentech/ avoiding cases of myocardial infarction. and the Veterans Affairs system in the
Investors and analysts have suggested Roche] or imatinib [Gleevec, Novartis]), United States, all of which have lower
that personalized medicine can minimize costly adverse events (such Payors’ adoption of personalized medicine membership turnover). Generating high-
dramatically reduce health care costs and as the warfarin dosing test), or delay tests is further complicated by the high quality health economic evidence will
help payors market products to the most expensive procedures can be extremely customer turnover experienced by provide the reimbursement confidence
attractive customers. Yet most payors cost effective for payors. Although such many commercial payors in the United that enables payors more rapidly to adopt
have been slow to invest in personalized tests cost between $100 and $3,000 States. This high turnover makes it tests and align physicians’ incentives with
medicine. Leaders in payor organizations each, they save $600 to $28,000 per less economically attractive for payors patient care and outcomes, rather than
identify several factors that could explain patient. By contrast, tests that save a to reimburse prophylactic tests that procedures. This could create a source
this reluctance. The first is an inability small amount per patient or have a low minimize the likelihood of conditions of competitive advantage for payors that
to identify easily which tests truly save probability of identifying patients requiring that will occur much later in life. Costs are more successful in identifying and
costs. The second is apprehension that intervention are not cost effective. For accrue to the payor that screens the implementing policies to promote cost-
it is difficult to track much of the earlier example, although BRCA1 testing to patient and performs the intervention, saving diagnostics.
stage/experimental testing, leading to predict the risk of breast cancer can save but the benefit accrues to the payor
fears that although individual tests may around $25,000 per patient identified, covering the patient when the disease Providers
not be that expensive, the overall eventual mutations are so rare in the general actually arises (perhaps ten years Today’s “procedure-based” reimbursement
costs could be unjustifiably high. A third population that this test, which costs up later). The pharmacoeconomics for the system for providers also presents a
concern is difficulty in enforcing standard to $3,000 per patient, is cost effective BRCA1 test illustrate the point (Exhibit challenge to the adoption of personalized
protocols to ensure that physicians follow only when performed on a patient with a 2). This longitudinal accounting issue is medicine. In this system, provider
through with appropriate patient care family history of breast cancer. Some tests particularly acute for diseases with a late economics will create incentives for
based on test results. Fourth, there is could also create costs on a per patient or delayed onset, whereby the insurer the use of some personalized medicine
potential for misuse of test for the elderly—for example, Centers for tests, but might discourage the use
information, particularly EXHIBIT 2 Medicare and Medicaid Services (CMS) of others. Physicians could be more
in the early stages of test The churn effect in the United States—accrues the benefit likely to embrace tests that increase the
Markers for prevention are the least cost effective for programs with high patient turnover
investigation/development, Lifetime cost savings for 10 patients with family Cost savings may not accrue to
of interventions that were paid for years number of procedures performed, while
which could lead to history of breast cancer screened for BRCA1 payors because of high patient churn
1
earlier by commercial payors. Notably, taking a more hesitant approach to those
patient harm. Fifth, there Plan types Lifetime Longest- ASO plans/
payor systems that have low patient that diminish procedure volume. For
2
36 Typical non-
care: term US consolidated ASO plans:
▪ UK plans: market: ▪ HMO3
is a lack of longitudinal 32 ▪ Europe ▪ Medicare
▪ Kaiser
▪ Most Blue
Cross and
turnover, such as integrated systems like example, a test that identifies patients at
accounting that would ▪ Long-term Blue Shield
employer plans Kaiser Permanente in the United States high risk of colon cancer such that they
▪ Mid-term
enable payors to capture -15
32
51 employers or single-payor systems in Europe, are require colonoscopies at three times
Average n/a 10 5 3
long-term cost savings number
years
of
BRCA1 only cost effective less exposed to this incentive challenge. the normal frequency would align well
for plans that capture most
from near-term testing. Cost to
screen
Cost of
inter-
Saved
lifetime
Saved
lifetime
Total
savings
patient
with plan of the lifetime value with gastroenterologists, given that the
patients vention cost of cost of with
with
family
breast
cancer
ovarian
cancer2
family
history
Savings/
loss per 10
36 10 -6 -20 As described above, personalized lifetime value of a patient related to such
history
To understand which patients,
$ thousands medicine tests span a spectrum from cost a molecular diagnostic is around $2,000.
tests actually save costs, Other tests may be cost neutral, or have
1
We estimated the lifetime cost savings for ten patients with a family history of breast cancer who are screened for BRCA1
variants associated with high risk of breast cancer. We assumed the total cost of screening ten patients is ~$15,000 (although effective to cost creating. Because the
the cost per patient can be as high as ~$3,000 according to quotes from the Myriad company web site). Twenty percent of
we analyzed various the cases were assumed to be positive and each of those two patients received an intervention consisting of a mastectomy
and a salpingoophorectomy (although standard of care for interventions varies), which together cost ~$16,000. Savings are
actual cost savings may not be known negative microeconomic incentives for
based on estimates of the lifetime costs of breast cancer and ovarian cancer drawn up by various agencies, such as the
types of test (see sidebar, California Breast Cancer Research Program. The figures were then applied to payors with different rates of member turnover
to calculate the savings. Approximate turnover rates are based on expert interviews as well as an analysis of internal data
until the test has been on the market their use. For example, Oncotype Dx, a
“Additional notes,” note 1). 2
from commercial payors.
Administrative services only. for some time, it will remain in payors’ gene-based diagnostic test for breast
3
Health maintenance organization.
90 Pharma R&D Compendium 2010 91
The microeconomics of personalized medicine

cancer patients that can be used to companies are moving slowly towards the EXHIBIT 4 incentives for companies
assess the likelihood of benefit from application of biomarkers and companion Scientific and commercial potential to pursue companion
Companies are likely to invest in areas with the highest potential rewards
1

chemotherapy, ultimately reduces the diagnostics. This is evidenced by the fact diagnostics, there are
number of patients that physicians that while the most aggressive players Scientific 2.8 significant potential
potential of
treat with such chemotherapy, and have biomarker programs for 100 percent companion
diagnostics
2.6 Oncology
Anti-infectives
commercial benefits from
2.4
thus the revenue that those patients and companion diagnostics for 30 percent 2.2
Antidepressants increased market share
Anticoagulants Autoimmune drugs
generate. Even so, Oncotype Dx has or more of their compounds, the average 2.0 CNS drugs Antipsychotics and pricing power. At
Statins
been widely adopted because of its company has far fewer (30 to 50 percent 1.8 Diagnostics
company activity Early pharmaceutical activity the same time, there is
1.6
clinical merit, but this example illustrates and less than 10 percent respectively). 1.4
No go Watchful waiting
also significant risk, as
Proton Asthma
the challenges that such tests can Moreover, many of the experts we 1.2 pump
inhibitors
Antihypertensives
Diabetes companion diagnostics
pose to providers’ economics. interviewed stated that their corporations 1.0
Antiarrhythmics
Antihistamines Growth factors
divide the treatable
0.8
had not prioritized companion diagnostics 0.6
Anti-inflammatories
patient population into
Pharmaceutical and and were taking a “cautious approach” 0.4 sub-segments and can
0 1.4 1.5 1.6 1.7 1.8 1.9 2.5 2.6 2.7
biotechnology companies to investments. Scientific and clinical Economic potential of companion diagnostics
reduce market share in
These companies are now using factors pose some limitations to the pace 1
some cases. Given this,
Rank order estimates for the scientific and commercial potential of the development of companion diagnostics in various
biomarkers to aid R&D, and in some of development. In some disease areas, therapeutic areas based on both qualitative factors (such as expert interviews), and quantitative factors (such as data on
price premiums for drugs launched in the same therapeutic class). Results should be taken as directional only.
companion diagnostics
cases will develop these markers as understanding of molecular mechanisms is are most likely to be value
companion diagnostics (tests to identify insufficient to select biomarkers rationally creating for later-to-market
patients’ likelihood of responding to at early stages of development. In other productivity. Indeed, it seems companion entrants in crowded markets characterized
a drug or experiencing side effects). areas, there is not a large clinical need for diagnostics may do little to improve by significant pricing flexibility.
R&D executives at 16 of the top 20 companion diagnostics. However, in many development productivity. In many cases,
biopharmaceutical companies interviewed disease areas, companies are moving they might actually increase overall cost For example, if two drugs are already
in a survey by McKinsey in 2007 indicated slowly despite scientific advances. and delay development. With respect on the market and are relatively
that, on average, 30 to 50 percent of to clinical trials, experts suggested that undifferentiated, the third drug on the
drugs in development have an associated Our research suggests that the potential Phase II trials often have to be larger when market is likely to capture a relatively
biomarker program, and suggested this to generate greater value after marketing companion diagnostics are employed. In small share—say, 5 to 20 percent (see
number was likely to increase. By contrast, through increasing price and market share practice, trials often need to be designed “Additional notes,” note 4). A companion
the same executives also suggested is vastly more important for the economics with several potential candidate biomarkers diagnostic that identifies a segment of
that fewer than 10 percent of drugs with of pharmaceutical and biotechnology in Phase II (and sometimes Phase III) as it the patient population that will respond
current biomarker programs would be companies than improving development is unclear which markers will be predictive especially well to a drug or have lower
launched with a companion (see “Additional notes,” note 2). In addition, toxicity, and thereby enables higher
diagnostic over the next EXHIBIT 3 the US Food and Drug Administration (FDA) pricing, could generate value. A key
five to ten years (and Where the commercial value lies is likely to require that “marker-negative” determinant of pricing diversity is payors’
Companion diagnostics could have most impact on pricing and market share
this is highly dependent patients be included in Phase III trials, price scrutiny/sensitivity, which varies
on the disease area). based on concerns that the drug could dramatically by disease area, particularly
Impact per new molecular entity developed with a
Value lever companion diagnostic,1 $ million be used off-label in these patients. This in the United States. This is illustrated by
In theory, companion Pre-market Avoiding recalls 0 20 practice is likely to eliminate the upside from Bidil, a fixed-dose combination of two
diagnostics can improve smaller trials that has been widely cited in generic cardiovascular drugs, hydralazine
Trial size -180 250
R&D productivity by recent years (see “Additional notes,” note and isosorbite dinitrate, that has been
decreasing trial size, Attrition rate 11 160 3). Apart from trial size, other commonly approved by the FDA specifically for
reducing attrition, and/or cited applications of personalized medicine African Americans with heart failure. In
increasing speed to market, Speed to market -330 680
during drug development also seem unlikely this case, attempts to charge a price
and enhance commercial to improve drug development productivity premium were met with aggressive
Post-market Price 170 ~1,700
performance by boosting substantially (Exhibit 3). differential co-pay tiering by payors,
market share and/or Market share -983 ~2,300+ which contributed to lower sales than
supporting higher drug Although increasing development expected (see “Additional notes,” note
prices. However, many 1
High-end and low-end estimates for the impact of a companion diagnostics program were based on case examples and productivity may not provide sufficient 5). In therapeutic classes where payors
expert interviews.
92 Pharma R&D Compendium 2010 93
The microeconomics of personalized medicine

scrutinize prices less EXHIBIT 5 molecular diagnostic compared with is likely to increase the time to market by
intensely (oncology drugs, The business risks of molecular diagnostics 30 to 50 percent for most typical large at least a year (see “Additional notes,”
Sensitivity analysis for factors affecting the commercial potential of a company developing a molecular diagnostic1
for example), companies laboratory companies [see “Additional note 8). That said, good communication
would be more likely to Sensitivity assumptions notes,” note 6]). Indeed, a number of between the Center for Drug Evaluation
charge a premium and Worst
case Today
Best
case
Impact on 10-year NPV of EBITDA2,
$ million
emerging companies, including Genomic and Research and the Office of In Vitro
maintain coverage. 1 Cost to develop test, $ million 50 45 40 -10.0 5.8
Health, Myriad, Celera, Monogram, Diagnostic and Device Evaluation and
and Xdx, have been successful Safety may partially mitigate this for
2 Timing of test Timing of development 6 5 4.5 -24.5 19
These companies should approval and
adoption
and approval, years in raising funding and developing priority reviews. It remains unclear what
be, and are, considering Time to payor
coverage, years
6 6 1 0 30
innovative molecular diagnostic tests. the approval timelines for other systems
investing in personalized Time to physician
adoption, years
8 8 5 0 15 will be. The European Medicines Agency
medicine in certain disease 3 Peak sales price 2,500 3,000 3,500 -7.7 7.7
Unfortunately, the molecular diagnostics (EMEA) and Japan’s Pharmaceuticals and
areas. To highlight disease Base case 10-year
business case still holds significant risk Medical Devices Agency (PDMA) also have
areas where near-term NPV = 14.8
(Exhibit 5). A number of factors contribute yet to establish clear guidelines for how
1
A representative profit and loss (P&L) model for a start-up molecular diagnostics company was created from a number of
investment in companion sources. The aim of this model was not to define the P&L statement for all such molecular diagnostics companies, but to to this risk, including development costs, they will treat the approval of personalized
create a model that would allow us systematically to explore the factors affecting profitability. The cost of test development
diagnostics is most likely (including investments in start-up infrastructure) was based on interviews with venture capital groups and start-ups as well as
actual data on seed funding for relevant companies. To assess the impact of various factors, we used estimates from expert
timing of development and approval, time to medicine tests.
to occur, we segmented interviews as well as historical data.
Earnings before interest, taxes, depreciation, and amortization.
2
payor coverage, rate of provider adoption,
drug classes according and peak sales price. To understand the With respect to payors’ adoption, the case
to their scientific and relative importance of these factors, we of Oncotype Dx demonstrates the challenge
commercial potential (Exhibit 4). This to justify approval, reimbursement, and modeled the economics of a hypothetical of slow coverage. Although the test was
segmentation is based on quantitative price. With this in mind, companies should start-up esoteric diagnostics company, launched in 2004, analysts and company
factors, as well as qualitative factors from act quickly to build the capabilities and then performed a sensitivity analysis using estimates suggest it will be 2010 before all
interviews, and the results should be experience that will be needed to succeed. upside and downside scenarios for each payors routinely cover the test. Coverage
taken in this light. Our analysis indicates variable. It should be noted that this model stands at about 85 percent. This contrasts
that companies are most likely to invest Diagnostics companies was based on benchmarks from a few starkly with typical adoption of a new
in diagnostics in areas such as oncology, Diagnostics and life science tools molecular diagnostics businesses with drug. In the United States, new drugs are
immunology, and infectious disease. The companies enable a wide variety of test the intent of testing the importance of risk generally reimbursed immediately at launch
segmentation also reveals disease areas types, including companion diagnostics factors. The model does not represent a or within the year. In Europe, drug coverage
where technical feasibility and clinical (often in collaboration with a biotechnology specific company, and the economics for may take slightly longer, depending on the
need exist, but where incentives are not or pharmaceutical company), early-stage companies with products currently on the extent of the review, but is unlikely to take
aligned to drive investment. These areas, diagnostics, disease recurrence and market vary significantly. Based on this more than four years—the adoption timeline
such as anticoagulants, antipsychotics, monitoring tests, adverse drug events model, the expected ten-year net present experienced by Oncotype Dx.
and antidepressants, are ripe for tests, and genotypic risk marker analyses. value (NPV) for an average diagnostic test
development by other organizations, such However, diagnostic test developers have is around $15 million. The most important Start-up diagnostics companies therefore
as diagnostics companies. faced difficulty capturing the full value factors influencing profitability are the time face challenging economics. However,
they generate, as exemplified by the fact to approval and rate of payor adoption. If as more tests become available and
However, companies should also realize that diagnostic tests are estimated to the time to approval is delayed by a year, payors, regulators, and molecular
that the payor environment is evolving influence between 60 and 70 percent the ten-year NPV becomes negative at diagnostics companies gain experience,
rapidly and application of personalized of all treatment decisions, yet account around -$10 million. This finding is relevant development and adoption times are likely
medicine tools will increasingly be for only 5 percent of hospital costs and given that it remains unclear how the FDA to shorten. Likewise, as the regulatory
required to preserve value. Although 2 percent of Medicare expenditure.4 will regulate in vitro diagnostic multivariate process becomes clearer—but potentially
pharmaceutical and biotechnology Molecular diagnostics are often cited as index assays (IVD-MIA). At the time of longer—payors’ adoption rates may
companies need to be aware of areas a more attractive market segment than writing, the FDA suggested a 510(k) may also increase. Given payors’ trepidation
where diagnostics can destroy value by typical diagnostics, given the potential be sufficient for tests which are prognostic about personalized medicine testing, it
sub-segmenting their existing markets, it for higher prices ($100 to $3,000 per indicators (see “Additional notes,” note 7), will therefore be advantageous for leading
will be equally important to prepare for an test compared with $20 to $50 for a but a premarket approval (PMA) from the diagnostics companies to help shape
environment in which regulatory bodies will typical diagnostic test) and higher gross FDA is likely to be required if the test directly the development of rigorous but efficient
demand greater proof of patient outcomes margins (50 to 70 percent for a sample influences therapy decisions. PMA review regulatory and approval standards.
94 Pharma R&D Compendium 2010 95
The microeconomics of personalized medicine

Potential catalysts for for personalized medicine tests, especially Research details
personalized medicine laboratory-developed tests, to be used in
To gather stakeholders’ perspectives on personalized medicine we conducted 60 interviews in the
therapy decisions?
first half of 2008 with executives and opinion leaders from leading private payor organizations,
We have described how current market
academic research institutions, health care provider organizations (such as academic medical centers
failures limit the speed of adoption of For the new regulations under
and hospitals), regulatory bodies, biopharmaceutical companies, molecular diagnostics and clinical
personalized medicine, and how solutions consideration, authorities need to weigh
laboratory companies, and venture capital funds. The interviewees were:
to these economic challenges represent short-term costs against long-term
• Eight payor executives including individuals from private payors (for example, Blue Cross/Blue Shield
opportunities to accelerate market benefits. Current plans include classifying
and Health Net) and CMS. The expertise of these individuals spanned coverage decisions and health
development. On the basis of conversations tests as Class I, II, or III, based upon the
technology assessment.
and analyses conducted during the course level of risk of the intended use. IVD-MIA
• Twenty biopharmaceutical executives in positions ranging from vice-president to chief executive
of this investigation, we see four main changes that promote more rigorous
officer. Expertise among these individuals spanned business strategy and operations, R&D, regulatory
catalysts that could significantly affect the evaluation of safety and effectiveness may
affairs, and reimbursement.
adoption of personalized medicine in the have long-term benefits by encouraging
• Thirteen diagnostics executives from large clinical laboratory companies and small and mid-sized
near term. faster adoption by payors and physicians
molecular diagnostics companies. All interviewees were senior executives with long experience of the
owing to the higher approval standards.
diagnostics industry.
1. Maximizing transparency and efficiency However, the near-term consequences
• Six researchers from leading academic institutions in the United States and the United Kingdom who
of the regulatory-approval process may harm short-term market investment.
are recognized as experts in molecular genetics, pharmacogenomics, bioinformatics, and molecular
and protein diagnostics.
2. Increasing the pace and predictability For diagnostics companies, the approval
• Three venture capitalists from leading firms that focus on molecular diagnostics investments.
of payor coverage for appropriate tests process can actually be an opportunity
• Two attorneys with legal expertise spanning intellectual property, FDA regulation, and health
to justify higher value/pricing by showing
care law.
3. Aligning reimbursement practices to willing to set appropriately stringent
• Eight regulatory experts from the Department of Health and Human Services, the FDA, and NICE.
incentivize appropriate diagnostic use by standards, and by shaping regulatory
physicians guidelines to bolster the industry and
We asked each interviewee’s opinions on the challenges and opportunities in personalized medicine for all
protect patients. For its part, the FDA
stakeholders, and discussed in more detail the use of personalized medicine in their own field of expertise
4. Encouraging pharmaceutical and should work to minimize approval delays
both currently and over the next five years.
biotechnology companies to take a long- that result from higher standards, and
Details of the quantitative analysis and financial modeling we conducted to understand specific
term investment view. help mitigate any negative impact on
stakeholder issues are highlighted in the exhibits.
investment in development. Leading
Regulatory environment pharmaceutical, biotechnology, and
First, regulatory bodies such as the FDA diagnostics companies should look for companion diagnostics on the basis of how this coordination will work in other
must improve the clarity and efficiency of opportunities to help shape the development retrospective tests of the Dx marker (that parts of the world and processes have
the test regulatory approval processes, of these guidelines and standards. is, performed on archived samples). Finally, not been established—for example, at
both for stand-alone and companion governments and regulatory bodies could the time of writing, the National Institute
diagnostics. These clarifications are critical To drive changes in market incentives, reward the development of companion for Health and Clinical Excellence (NICE)
to diagnostics companies’ ability to plan regulatory bodies such as the FDA diagnostics directly by increasing the in the United Kingdom had not reviewed
ahead and to design trials. Based on our and EMEA could decide not to require patent life for drugs developed with a molecular diagnostics test. State
conversations with more than 60 experts, collection of clinical data on marker- companion diagnostics, providing tax- payors, private payors, and diagnostics
the key questions that regulatory bodies negative patients, thus lowering based incentives, and continuing to award companies can help fuel growth in
such as the FDA and EMEA should address development costs. Concerns about grants for R&D. the personalized medicine market by
include: Will marker-negative patients the use of therapeutics in the “marker- coordinated efforts to improve the pace
be required for Phase III trials? Will use negative” population could be reduced Payor coverage and process for coverage decisions. One
of archived samples or “flexible” trial by parallel moves by payor organizations In the United States, approval and step could be for CMS to take a lead in
designs be permitted for approval of and regulatory bodies to increase barriers reimbursement coverage decisions aligning the reimbursement process with
companion diagnostics, and under to “off-label” use. Furthermore, regulatory represent two discrete processes with the regulatory approval process. Pre-
what circumstances? What regulatory bodies could increase the flexibility of trial minimal coordination between the FDA submission meetings to delineate data
standards and oversight will be required design and even allow for the approval of and CMS. Uncertainty remains about requirements for regulatory and coverage
96 Pharma R&D Compendium 2010 97
The microeconomics of personalized medicine

approval and ongoing joint reviews can predominate in Europe are at an advantage Investment by pharmaceutical and (Velcade) for multiple myeloma and the
facilitate interagency collaboration. Optimal when it comes to the adoption of biotechnology companies interferon-beta drugs for multiple sclerosis;
alignment across the two agencies would personalized medicine, in two ways. First, Pharmaceutical and biotechnology here reimbursement is contingent upon
imply that if suitably stringent guidelines they are not as susceptible to longitudinal companies should take a long-term patient outcomes. Similarly, payors could
are set, then CMS would provide coverage accounting issues. Second, coverage investment view. Some already do: create innovative risk-sharing agreements
and adequately reimburse those who decisions can be less complex and involve leaders we interviewed who have invested with diagnostics companies. A test
meet those hurdles. For example, the fewer decision makers. most heavily in personalized medicine could, say, receive conditional, partial
requirement of additional health economic suggested they are renewing their focus reimbursement for a number of years until
data and/or regulatory approval for clinical Physician incentives on outcomes and clinical value in the the clinical effectiveness was definitively
claims may be reasonable prerequisites Beyond improvements associated with process of drug discovery. They realize demonstrated (at which point the
for coverage, and could thus help ensure regulatory approval and formal coverage, that the drugs they are developing diagnostics company would be paid in full).
adequate reimbursement, pricing, and aligning physicians’ incentives could today will be entering markets with more The payor limits cost exposure by covering
value for diagnostics players. further hasten adoption. Reimbursement competitors, more pricing pressure, and part of the costs for a limited time, while
schemes in many countries remain a higher bar for differentiated clinical diagnostics companies benefit from early
Development of formal guidelines could largely “activity based,” with physicians outcomes. Not surprisingly, these same coverage decisions.
improve the transparency and efficiency receiving disproportionately higher rates companies are investing heavily in
of decisions relating to coverage. for procedure-oriented services than for personalized medicine. Over the next few decades, the
Today, CMS typically makes coverage evaluation and management activities. development of -omics sciences and
decisions for molecular diagnostics at As such, there is little financial incentive An aggressive move towards value- or supporting technologies will enable the
the regional rather than national level. for physicians to perform tests that might outcomes-based pricing by CMS or private creation of an increasing number of
As a consequence, decisions are made prevent the need for further treatment. payors could greatly increase the financial personalized medicine tests. However, the
many times based on different guidelines In fact, there may be a real financial value of personalized medicine and so use of these tests could be hampered by
and processes and often with differing disincentive. the incentive to invest in it. One possibility poorly aligned incentives for stakeholders.
outcomes (see “Additional notes,” note 9). might be to employ innovative risk-sharing All stakeholders should therefore work
Private payors also lack clear guidelines Efforts are under way to shift towards models for drug and diagnostic coverage. together to help reshape these incentive
for these decisions. Both CMS and a more “outcome-based” approach to For instance, payors could follow the structures and so reap the benefits of
private payors have an important role to reimbursement, a system that will provide examples in Europe of bortezomib personalized medicine.
play in shaping coverage and payment incentives for physicians to use and act
decisions. Private payors we interviewed on appropriate personalized medicine
A version of this article, entitled “The microeconomics of personalized medicine: today’s challenge
are waiting to understand and potentially diagnostics. Yet payors should also
and tomorrow’s promise,” was first published in Nature Reviews Drug Discovery, April 2009,
follow CMS coverage policies (as often work to develop a system that ensures
Volume 8, No 4, pp. 279–86. For further details of the analysis and methodology, please contact the
occurs with therapeutics). physicians are reimbursed for the test itself
authors.
in order to encourage adoption. Moreover,
One way to improve coverage guidelines personalized medicine tests today are
Jerel Davis (jerel_davis@mckinsey.com) is a consultant in McKinsey’s Silicon Valley office, where
in both systems and processes would billed in the United States by “CPT code
Saumya Sutaria (saumya_sutaria@mckinsey.com) and Laura Furstenthal
be to establish an agency to assess the stacking,” whereby a multivariate assay
(laura_furstenthal@mckinsey.com) are principals, and Edd Fleming (edd_fleming@mckinsey.com)
clinical and cost effectiveness of tests. This is billed by adding multiple, generic
and Philip Ma (philip_ma@mckinsey.com) directors. Amar Desai (amar_desai@mckinsey.com)
agency could be a coordinated effort by codes—for example, for a diagnostic
is a consultant in the Los Angeles office. Troy Norris (troy_norris@mckinsey.com) is a consultant
payors, CMS, interested pharmaceutical based on a single gene (see “Additional
in the New York office.
and biotechnology companies, and notes,” note 10). This approach is not
diagnostics players, and could take the scalable and can lead to billing practices
form of a third-party, non-profit agency, a in which laboratories game the system.
consortium, or a new government agency. Eventually, individual codes will need to be
The formation of new oversight agencies developed for each molecular diagnostic
(for example, an FDA center for diagnostics) that are commensurate with the cost and
could also help in this regard. Notably, value of the test and provide appropriate
single payor systems such as those that reimbursement to physicians.
98 Pharma R&D Compendium 2010 99
The microeconomics of personalized medicine

Additional notes

Note 1. Some payors noted that newer thyroid cancer diagnostics have led to a dramatic increase
in the incidence of thyroid cancer (a 250 percent increase from 1973 to 2002), but no improvements
in mortality.5 One explanation for the findings is that most of the incremental detection was for
papillary cancers with a very good prognosis.

Note 2. Experts we spoke to indicated that between two and six markers are typically chosen for a
drug’s companion diagnostic program. These markers are usually chosen before Phase II, developed
in parallel with the Phase II clinical trial, and then tested retrospectively on Phase II participants.

Note 3. A widely cited example in this respect is the Phase III trial for the anticancer drug
trastuzumab (Herceptin, Genentech/Roche), a monoclonal antibody that targets HER2. The trial
included only 470 breast cancer patients and only a marker-negative arm. The expected size of the
trial without a companion diagnostic based on HER2 expression levels has been estimated at 2,200
patients (based on a presentation by Arthur Levinson, CEO of Genentech, in October 2003).

Note 4. This figure is derived from McKinsey analysis and IMS sales data; the estimated range is
based on 5 percent average market share for third-to-market drugs at around one to three years
post launch, and 20 percent average market share of second-to-market drugs at around one to three
years post launch.

Note 5. Bidil was priced at a premium. In interviews with four different insurance companies,
payors indicated that they differentially tiered the drug because they did not think the clinical benefit
justified the cost.

Note 6. These figures are based on McKinsey analysis of selected molecular diagnostics and
traditional diagnostics.

Note 7. Section 510(k) of the US Food, Drug, and Cosmetics Act requires device manufacturers to
notify the FDA of their device at least 90 days in advance of marketing. The review process for this
Premarket Notification is more straightforward than Premarket Approval and typically takes less
than 90 days.

Note 8. These figures are based on historical approval times, and include non-direct review time and
direct review time. Premarket approval typically takes around 18 months whereas registration takes
some six months.6 1
Iakoubova, O. A. et al. “Association of the Trp719Arg polymorphism in kinesin-like protein 6 with
Note 9. The Centers for Medicare and Medicaid Services (CMS) may make national coverage myocardial infarction and coronary heart disease in 2 prospective trials.” J. Am. Coll. Card. 51,
determinations for certain molecular technologies, whereas coverage for most laboratory tests is 435-443 (2008).
determined locally by CMS. A local coverage determination is based on a review of current medical
2
Shiffman, D. A. et al. “Kinesin family member 6 variant is associated with coronary heart disease in
practices and clinical data, and procedures for coverage decision are not uniform across localities. the women’s health study,” J. Am. Coll. Card. 51, 444-448 (2008).
3
Iakoubova, O. A. et al. “Polymorphism in KIF6 gene and benefit from statins after acute coronary
Note 10. Reimbursement and billing for molecular diagnostics are performed using current syndromes.” J. Am. Coll. Card. 51, 449-455 (2008).
procedural terminology (CPT) codes. Most molecular diagnostics do not have a single unique code 4
Zafar, I., Levy, T., & Damergy, S. “Third Wave Technologies Report, Hospital Supplies and Medical
assigned. Billing for multivariate tests involves “stacking” multiple codes that describe individual Devices,” Deutsche Bank, May 12, 2008.
components of the assay. For example, billing for a single Myriad’s BRCA panel can involve 5
Davies, L. & Welch, H. G. “Increasing incidence of thyroid cancer in the United States, 1973–
employing five different codes and 171 separate CPT units. 2002,” JAMA 295, 2164–2167 (2006).
6
Goodman, C. et al. “The Value of Diagnostics, Innovation, Adoption, and Diffusion into Health
Care.” The Southern California Biomedical Council [online], http://www.socalbio.org/pdfs/
thevalueofdiagnostics.pdf (2005).
101

The authors of these articles represent a cross-section of McKinsey’s


Authors global R&D practice. We welcome your thoughts and reactions to the
ideas presented. Please email us at pharma_r&d@mckinsey.com or
contact the authors individually.1

Mark Beards (mark_beards@mckinsey.com) is a consultant in the London office


Eric David, MD (eric_david@mckinsey.com) is an associate principal in the New York office
Jerel Davis, PhD (jerel_davis@mckinsey.com) is a consultant in the Silicon Valley office
Áine Denari (aine_denari@mckinsey.com) is a consultant in the Dublin office
Amar Desai, MD (amar_desai@mckinsey.com) is a consultant in the Los Angeles office
Michael Edwards (michael_edwards@mckinsey.com) is an associate principal in the London office
Matthias Evers, PhD (matthias_evers@mckinsey.com) is a principal in the Hamburg office
Edd Fleming, MD (edd_fleming@mckinsey.com) is a director in the Silicon valley office
Laura Furstenthal, PhD (laura_furstenthal@mckinsey.com) is a principal in the Silicon valley office
Maria Gordian, MD (maria_gordian@mckinsey.com) is a principal in the New York office
Michele Holcomb, PhD (michele_holcomb@mckinsey.com) is a principal in the New Jersey office
Philip Ma, PhD (philip_ma@mckinsey.com) is a director in the Silicon Valley office
Sam Marwaha (sam_marwaha@mckinsey.com) is a director in the New York office
Martin Møller (martin_moller@mckinsey.com) is a principal in the Copenhagen office
Troy Norris (troy_norris@mckinsey.com) is a consultant in the New York office
Richa Pande, MBBS (richa_pande@mckinsey.com) is a consultant in the New Jersey office
Paul Rutten (paul_rutten@mckinsey.com) is an associate principal in the Amsterdam office
Mubasher Sheikh, MD (mubasher_a_sheikh@mckinsey.com) is a principal in the London office
Navjot Singh, PhD (navjot_singh@mckinsey.com) is a principal in the New York office
Saumya Sutaria, MD (saumya_sutaria@mckinsey.com) is a principal in the Silicon Valley office
Tony Tramontin, PhD (tony_tramontin@mckinsey.com) is an associate principal in the New York office
Rodney Zemmel, PhD (rodney_zemmel@mckinsey.com) is a director in the New York office

McKinsey & Company is a global consultancy firm that helps leading corporations and organizations
to make distinctive, lasting, and substantial improvements to their performance. McKinsey advises
companies on strategic, operational, organizational, and technological issues. Of our 8,000 consultants
worldwide, more than 1,600 focus on health care, and 30 percent of these hold PhD or MD degrees.
In the past four years, we have completed almost 4,000 engagements in health care, spanning all
major sectors and functions of the industry.

1
This list includes current McKinsey personnel only.
Copyright © McKinsey & Company
Pharmaceutical and Medical Products R&D Practice
2010

Das könnte Ihnen auch gefallen