Sei sulla pagina 1di 48

From Molecule

to Medicine.
Delivered.
CONTENTS
 BIOLOGIC DRUG SUBSTANCE
4 Managing Demand Uncertainty in Biologics Production
8 Is Your In-House Strategy Ready for the Uncertainties of Biologic Drug Development?

 API DRUG SUBSTANCE


11 How Can You Avoid the Fallout from Incompatibility Between Your API and its Formulation?
13 Trends and Challenges in Outsourced Oral Solid Dosage Forms: A Q&A

 FORMULATION DEVELOPMENT
15 Characterizing Drug Substance Properties Early Can Optimize Drug Product Formulation

 PHARMACEUTICAL PROCESS DEVELOPMENT
18 Avoid the Do-Over: Why Early Investment in a Scalable Manufacturing Process Is Critical
22 Solving the OOS Problem with Continuous Manufacturing

 PATHEON ONESOURCE™
25 Single-Vendor CDMOs Bring Speed and Cost Savings to the Table
28 4 Perspectives on Single-Vendor Drug Development Models
28 Bringing the Single-Vendor Approach to the Pharma Industry
29 New Resource Saves Drug Developers Time and Money
31 A Single-Vendor CDMO Eases Communication Flow
32 Smaller Biotech Companies and the Single-Source CDMO

 CLINICAL SERVICES
34 Cold-Chain Fully Automated Assembly and Labeling of Pre-Filled Syringes for Clinical Trials

 COMMERCIAL MANUFACTURING AND TECHNOLOGY TRANSFER


36 Considerations in Selecting an Outsourced Solution
38 Technology Transfers: Reaping Rewards, Reducing Risks

 CLIENT SUCCESS STORIES


41 How Pacira Pharma is Working to Help Curb the Opioid Epidemic
43 Fighting the Opioid Epidemic: How Grünenthal’s Abuse-Deterrent Technology Contributes

2 www.patheon.com ● doingbusiness@patheon.com
From the
President

Expanding Capabilities
Ongoing evolution in the industry global reach in clinical trials packaging, labeling, distribution and
logistics services takes clients from drug formulation to clinical
We need to look no further than recent headlines to see trials support to commercial production seamlessly. The
evidence of the exponential changes and innovations happening combination also benefits biotech customers with the availability
today in the pharmaceutical industry. The first gene therapy of state-of-the-art bioproduction technologies and biologics
was approved in July 2017 by the U.S. Food and Drug development and production expertise to shepherd new biologic
Administration (FDA) following the approval of other gene therapies throughout the development timeline.
therapies for rare diseases in Europe. These targeted therapies
have the potential to significantly change the industry and most It is an exciting time in the life sciences industry. The Thermo Fisher
importantly improve the lives of patients. Pharmaceutical and and Patheon combination is well positioned to help clients
biotechnology companies are constantly seeking to better accelerate innovation, drive productivity and deliver on our mission
understand diseases and develop better therapies to meet the to enable our clients to make the world healthier, cleaner and safer.
growing needs of patients around the world.
”From Molecule to Medicine” is a compilation of insights and
These technological advances and novel compounds are perspectives from Patheon’s scientific, manufacturing and
increasingly complex, requiring specialized resources, scientific engineering experts. We selected articles to provide a view
expertise and advanced manufacturing technologies to produce. across the industry — from small and emerging biopharma
More and more companies are forging strategic relationships starting on the drug development journey to scale-up,
with external partners to create customized solutions and deliver technology transfer, and commercialization. I hope you find its
technical expertise to accelerate delivery of new therapies to contents valuable.
market. Strong partnerships enable pharma and biotech
companies to focus on research and commercialization. As the
industry creates these new advances and approaches to treating
diseases, so too must the contract drug development and
manufacturing companies.

Combining resources to deliver value, from


molecule to medicine
With Thermo Fisher Scientific’s recent acquisition of Patheon,
the company is part of an organization dedicated to making the
world a healthier place. It brings an end-to-end solution designed Michel L agarde, President
to accelerate drug development and simplify our clients’ supply
chains. Patheon’s highly complementary capabilities in drug
substance, pharmaceutical development and drug product
manufacturing combined with Thermo Fisher’s expertise and

Patheon’s highly complementary capabilities in drug substance, pharmaceutical development and drug product
manufacturing combined with Thermo Fisher’s expertise and global reach in clinical trials packaging, labeling, distribution
and logistics services takes clients from drug formulation to clinical trials support to commercial production seamlessly.

www.patheon.com ● doingbusiness@patheon.com 3
Biologic
Drug Substance

Managing
Demand
Uncertainty
in Biologics
Production

Stephen Lam John Ward


Senior Vice President, Vice President,
Head of Biologics Engineering
When a biologics company prepares to launch a new product, it to make costs non-linear with volume. For example, say a man-
must forecast the manufacturing capacity it will need. To create ufacturer makes a 2,000-liter subculture batch, at a cost of
this forecast, it must factor in its estimate of the size of future about $1.5 million to $2 million (including raw materials). For a
sales, the timing of the launch, the dosage of the product, its titer of 1 gram per liter, that’s about 1.6 kilos of active pharma-
strategy for building its market and a host of other variables. Vari- ceutical ingredient (API), with yield losses, at a cost of $1,250 a
ations in any one of those factors can lead to drastically different gram. A 20,000-liter batch, which would cost $4 million, would
demand scenarios. If a company overestimates demand, it may yield about 16 kilos of product at $250 a gram. That’s 80% less
end up investing in too much capacity, and therefore find itself than the 2,000-liter batch. Much more cost effective.
paying more per unit of the product than it needs to, thus impact-
ing its margins. If it underestimates demand, it risks not being However, at the 20,000-liter batch size, the sponsor could end
able to satisfy demand, therefore losing revenue. up with too much product, some of which may expire before it
can be sold. That’s a loss. Further, the commitment to large-
Forecasting demand is a complex endeavor. For instance, it’s scale capacity is very expensive, whether in-house or via a
not unusual for the forecasted and actual dosage of a product
contract development and manufacturing organization (CDMO).
to vary by a factor of as much as three. Obviously, that makes a
Large-scale plants cost hundreds of millions of dollars, and
big difference to a demand forecast. If a manufacturer has built
many CDMOs require long-term commitments for large scale.
capacity in anticipation of a new product and its clinical trial is
delayed (for any number of reasons), that manufacturer’s capi- On the other hand, the manufacturer could run a batch only
tal is tied up in a fallow facility. For a small company for which every few years at large scale to guard against producing too
liquidity is critical, that can be catastrophic. much product. However, that might create scheduling issues
and degrade the effectiveness of the manufacturing organiza-
Why forecasting is so hard tion; change overs in large-scale plants can be very expensive.
The manufacturer also risks losing inventory, not just due to
When planning for capacity, a manufacturer must consider both product expiration, but also due to latent defects, issues that
volume and scale. Both are important, the effect of scale being may only become apparent after several years. Plus, one would

4 www.patheon.com ● doingbusiness@patheon.com
still need three to five validation batches, which being large,
would be expensive.
It’s in the middle — between
Of course, at a smaller scale of production, each unit costs more.
small and large scale — where
Large or small, the approach one chooses will have a ripple things get more complicated.
effect throughout the business, affecting hiring decisions, cash
flow, schedule duration, available capacity, cost of goods and
them from meeting demand, and they lose revenue. Worse, it
on and on. And the scarcity of outsourceable capacity at cer-
can lead to a permanent loss of market share to competitors
tain scales (for example, 20,000-liters) further complicates the
whose products may not be covered by their patent.
process — all of which sometimes leads companies to choose
a solution that is suboptimal. The cost per gram of your API is a good place to start planning
your capacity as it tends to be forecast-independent. It’s pri-
marily dependent on the titer, which you will know early on.
An Rx for navigating complexity
Armed with that, you can determine your cost per gram of the
It is possible though to mitigate forecasting risk so long as you API and check that it fits the projected price. If it does, your next
start the process early in the product commercialization step is to choose the manufacturing scale.
lifecycle. Here is our approach:

2. Choose the manufacturing scale


1. Determine a target cost per gram for the API
If you are a large manufacturer, you’ll generally have the option
Too often, this conversation does not happen until it is too late. to be in the $100 to $150 per gram band (See Exhibit 1), and
Companies fearful of overproducing often launch without you can use capacity you already have for large-molecule pro-
enough capacity. Then, post-launch, they must scale up, and duction. This will enable you to maintain a low cost and manage
they get caught in a long pre-approval process. That precludes your risk. For a large company with a broad product portfolio,

Client Cost ($/g)


700

600

500

400

300

200

100

0
42 105 210 315 420 525 1260 2520 3780 kg/Year
10 25 50 75 100 125 300 600 900 2000 L Batches
10 20 30 40 50 120 240 360 5000 L Batches
13 17 40 80 120 15000 L Batches

2-6 2000 L Own ($/g) 2000 L Outsource ($/g) 2 x 5000 L Own ($/g) 5000 L Outsource ($/g)

6 x 5000 L Own ($/g) 6 x 15000 L Own ($/g) 6 x 15000 L Outsource ($/g)

Exhibit 1: Scale, utilization and throughput have a significant impact on cost (based on a titer of 3g/l @ 75% yield)

www.patheon.com ● doingbusiness@patheon.com 5
Client Cost ($MM/Yr)
1400 Need big tanks

1200

1000
Differential
800
Many options are creates
viable in the “middle” driver for
change
600

400
2000 L scale
200
very viable

0
42 105 210 315 420 525 1260 2520 3780 kg/Year
10 25 50 75 100 125 300 600 900 2000 L Batches
10 20 30 40 50 120 240 360 5000 L Batches
13 17 40 80 120 15000 L Batches

2-6 2000 L Own ($/g) 2000 L Outsource ($/g) 2 x 5000 L Own ($/g) 5000 L Outsource ($/g)

6 x 5000 L Own ($/g) 6 x 15000 L Own ($/g) 6 x 15000 L Outsource ($/g)

Exhibit 2: Total cost expended identifies driving force for change

dedicated capacity allows it to flex to accommodate demand process, or both. You can always improve the process in paral-
variations across the portfolio and to build its expertise in large- lel and implement the new one when it’s ready.
molecule production.
In Exhibit 2, the vertical gap between the lines is the financial
For smaller production volumes, you will be on the left of the driver for change. For instance, at 1,000 kilos per year, that’s
Manufacturing Scale chart, and the cost will be about $350 per about a $300 million incentive to switch from 2,000-liter out-
gram if you outsource two to three 2,000-liter bioreactors. source to 15,000-liter outsource.

It’s in the middle, between small and large, where things get
more complicated. 3. Determine campaign size and frequency
For example, at 100 kilos per year, the cost would be about Plan the campaign to meet the expected annual demand and make
$35 million for an outsourced 2,000-liter process, versus about sure this will scale up at launch to meet peak projected sales. If the
$17 million for self-built. So, the answer could be to self-build. campaign volume is in the range, but your cost per unit is not, con-
sider how you might improve the yield. Conversely, if the volume is
Alternatively, the solution might be to develop the process to
improve the yield. That might cost say $20 million with all the too small, then consider a smaller scale for product launch, and
development work and regulatory refiling required. Improving either how to scale up post-launch or how to improve the yield.
the process often makes sense once the volume is in the range
of 20-50 batches per year. Then the lower API cost can often
pay back the initial investment within a few years while releas-
We recommend a
ing capacity for other products. Process development is often a
lot less expensive than building a plant, although the outcome
manufacturing strategy that can
is less certain. accommodate a forecast range
Or the right answer might be to outsource at first and then, if the and that you can adapt as
volume meets or exceeds expectations, look at the cost-benefit
of changing from outsourced to insourced, or of improving the better data becomes available.
6 www.patheon.com ● doingbusiness@patheon.com
4. Build flexibility into the scale
Clone Selection
One way to achieve flexibility is through multiplexing. For exam- 5 Clones
Selection based on growth, productivity, stability and product
ple, one company recently validated its process in 2,000 liters
stainless. It intends to obtain full approval for a multiplexed ar-
Process Development
rangement of six times 2,000 liters via a license variation. Parameters Studied: pH, temperature, perfusion rate, etc.

Another way to acquire flexibility is to combine solutions. For


Media Evaluation
example, you could outsource to a CDMO while working on Custom Media Optimization

improving the process or combine outsourcing with production


from your own large tanks. In fact, in the early stages, it is
Process Characterization & Scale-Up
sometimes a good idea to make capacity variable (with, for
example, several 2,000-liter tanks), and then consolidate as
forecasts become more certain. Combining multiplexing with a
Titer
future increase in downstream scale may simplify future tech-
nology transfers and regulatory strategies. By engaging in
these discussions earlier in the commercialization timeline, you
can develop multiple scenarios and test them against financial,
regulatory and commercial success factors. You can also use Clone
this time to discuss your potential strategies with regulatory Development

agencies during the clinical development process. Media


Scale-Up

5. Improve the yield


There are various new ways to improve API yield. One client
asked Patheon to conduct a full perfusion development pro-
gram on a complex recombinant protein to lower their cost of
Exhibit 3: Improvement in titer due process development
production. We went back to the cloning step and selected the
best of five options. We then tested 20 media combinations and
created an inexpensive custom medium. And then we scaled
and process yield. That means you can think about what cost
up the process. The result was a 6-fold improvement in titer and
band is optimal for each demand scenario and how best to
a 10-fold improvement in volumetric reactor productivity. You
maximize margins while reducing risks.
can see the improvement due to each step in Exhibit 3.
The best advice is to start early, build options into your process,
understand the lead times to make the changes and continue to
The early planner contains the uncertainty
evaluate your strategy during the commercialization process. In
Ask any planner, and they will tell you that the only thing they this way, you can reduce the uncertainty in your plans. ■
know for sure about the forecast is that it will be wrong. The
question is by how much. We recommend that you create a
manufacturing strategy that can accommodate a forecast range
and that you can adapt as better data becomes available.

Biologics manufacturers have many potential paths to accom-


modate demand forecast uncertainty, but once one is chosen,
it can be difficult (and expensive) to switch. The earlier you start
to plan, the more time you have to arrive at an option that’s best
for your company. Even outsourcing can have lengthy lead
times, even longer if the process is complex and the desired
volumes are high.

There is no reason not to begin planning early. By the end of


Phase I, a company will have a good idea of the dosing range

www.patheon.com ● doingbusiness@patheon.com 7
Biologic
Drug Substance

Is Your In-House
Strategy Ready
for the
Uncertainties of
Biologic Drug
Development?

John Ward
Vice President,
Engineering

The pharmaceutical industry’s past reliance on blockbuster drugs testing are approved by the FDA.4 This is because drug develop-
has evolved to include a focus on developing drugs that treat the ment is complex and unpredictable, especially for biologics, which
unmet needs of smaller patient populations. These niche drugs are developed by growing “live” cells in a bioreactor. Depending on
most often come in the form of biologics, which are an increasing- the conditions, the results can vary with each batch. These char-
ly larger share of new drug approvals in the past decade, from a acteristics make large-molecule drugs much more challenging to
low of 10% to a high of 27%.1 By 2022, 50% of the value of the top bring to market than small-molecule drugs. It also makes them
100 products is expected to come from biologics.2 more exciting, as the knowledge and expertise industry stands to
gain from this new frontier could potentially change the face of
This proliferating number of targets has had a profound effect on medicine; however, a company has to be able to withstand the
the complexity and cost of drug development, which can take up highs and lows of biologics development.
to 15 years to complete and, as outlined in recent estimates, can
cost up to $2.6 billion.3 Yet, despite the time and resources invest-
ed, only about 12% of the drug candidates that make it into Phase I Is it possible to achieve
efficiency and flexibility within
1 BioWorld, Biologics Share Of Medicine Chest Grows, And So Do Pricing
Concerns — http://www. bioworld.com/content/biologics-share-medicine-
chest-grows-and-so-do-pricing-concerns a drug developer’s own
2 EvaluatePharma, World Preview 2016, Outlook to 2022 — http://info.
evaluategroup.com/rs/607-YGS-364/images/wp16.pdf walls, or is outsourcing
3 Tufts Center for the Study of Drug Development, Tufts CSDD Assessment of
Cost to Develop and Win Marketing Approval for a New Drug Now Published
pharma’s best not-so-secret
weapon against today’s
— http://csdd.tufts.edu/news/complete_ story/tufts_csdd_rd_cost_study_
now_published

development uncertainties?
4 PhRMA, 2015 Profile, Biopharmaceutical Research Industry — http://www.
phrma.org/sites/default/files/pdf/2015_phrma_profile.pdf

8 www.patheon.com ● doingbusiness@patheon.com
To avoid costly mistakes and wasted efforts, a company must
prepare for any surprises along the drug development pathway.
One way to accomplish this is to diversify its strategies rather
than develop only a single-pronged attack for launching a prod-
uct. This flexibility creates the opportunity to incorporate multi-
ple solutions if necessary. By adopting an operational approach
that minimizes risk and optimizes drug development, a drug
developer can successfully manage its product portfolio and
reach its commercial goals. But is it possible to achieve effi-
ciency and flexibility within a drug developer’s own walls, or is
outsourcing pharma’s best not-so-secret weapon against to-
day’s development uncertainties?

Below are two main areas where a drug developer can face sensible route. As your throughput increases, the economics
significant obstacles during biologics development. By evaluat- start to shift to stainless steel, as the cost of SUT components
ing these capabilities, a drug developer can answer the critical can be expensive in a high-demand scenario. Without the abil-
question of whether its in-house strategy is ready for the uncer- ity to accurately predict demand, though, there is no way to re-
tainties of drug development. ally know which facility type is best for your product.

Considering the substantial costs of producing and storing bio-


Capacity: Is your facility prepared for the logics, the implications of an inaccurate forecast can be
demands of today’s biologics pipeline? far-reaching. When demand is overestimated, companies are
left with unused product and the likelihood of a significant loss
As part of pharma’s evolution, the demand for capacity in bio- in unproductive investments. This affects the company, its em-
pharma is changing. An increased focus on process technology ployees and its stakeholders. If it is underestimated, a manu-
improvements in pharma over the last 20 years has resulted in facturer is left scrambling to come up with enough product to
much higher yields, enabling pharma to reduce its programs and meet demand and creates new opportunities for its competi-
products to smaller capacities. These new production processes tion. As it attempts to come up with the needed capacity, the
and capabilities, such as perfusion and continuous processing, upfront costs of building infrastructure, hiring personnel and
are driven not just by the need for smaller batches but also by the installing the necessary systems begin to mount. Meanwhile,
complexity and instability of biologics. Other operational capabil- patients in need of lifesaving drugs are left waiting for their
ities like single-use technology (SUT) and online buffer medication. Still, the final product must be delivered quickly, at
conditioning offer additional major benefits, such as simplified a reasonable cost and at the highest quality possible.
logistics. While these technologies have become more sophisti-
cated, they can still be difficult to apply in production. Until these
technologies further advance, pharma manufacturers must Experience: “The better you putt,
come up with a more feasible plan to achieve flexibility and effi- the bolder you play.”
ciency in this new manufacturing landscape. This is especially
important, as a recent study by the American Pharmaceutical American golfer Don January once said the quote above to de-
Review shows that half of all products projected in the future can scribe the risks a golfer is willing to take with their shot if they
likely be met with a 5,000-liter bioreactor or smaller per product.5 know they have the skills to face — and prevail over — a com-
Fortunately, the emergence of SUT has brought promise to man- plicated situation on the green. Putting is difficult, and improving
ufacturers that need to quickly expand capacity and have the at it requires a lot of practice. The same can be said for devel-
resources to invest into this transition. SUT offers the flexibility oping biologics. A drug developer that is confident its team has
needed for bio’s future, and with facilities taking 12 to 18 months the necessary skills to overcome the challenges of biologics
to build (as opposed to the timeline for stainless-steel facilities of development is more likely to keep its business in-house. But a
five to six years for large-scale), it is an attractive solution to a small to midsize innovator company usually does not have the
growing need for capacity in a shorter period of time.
5 American Pharmaceutical Review, Global Biomanufacturing Trends,
Nevertheless, a manufacturer cannot take full advantage of the Capacity, and Technology Drivers: Industry Biomanufacturing Capacity
Overview — http://www. americanpharmaceuticalreview.com/Featured-
benefits of any facility type if the demand forecast it relies on is Articles/188840-Global-Biomanufacturing-Trends-Capacity-and-
Technology-Drivers-Industry-Biomanufacturing-Capacity-Overview
inaccurate. In lower-throughput scenarios, SUT is often a more

www.patheon.com ● doingbusiness@patheon.com 9
resources to staff a large team of experts. Therefore, it be- not having access to necessary expertise. This makes it impera-
comes unlikely they have the resources and experience not tive its team has the know-how to work with biologics.
only to foresee potential issues but also to quickly come up with
a solution that does not have a long-term impact on a product’s That expertise also extends to the regulatory side. More pro-
timeline and development costs. cesses moving through a plant results in more agency inspec-
tions. This builds a stronger, broader understanding of the
For example, technology transfers are a critical step and nec- regulatory framework as well as a familiarity of the agency with
essary even in an in-house scenario, as multiple sites separating the company itself, which can facilitate and even expedite a prod-
process development and manufacturing can exist. According uct’s speed to market. In addition, a drug developer must be able
to the ICH Q10 guidance6, “The goal of technology transfer ac- to recognize the impact of changes that can occur during devel-
tivities is to transfer product and process knowledge between opment and how this affects the final formulation (and, thus, its
development and manufacturing, and within or between manu- approval). For example, what happens when a drug’s intended
facturing sites to achieve product realization. This knowledge delivery system is initially an auto injector, but the concentration
forms the basis for the manufacturing process, control strategy, ultimately requires a dose higher than the 3-milliliter max of that
process validation approach and ongoing continual improve- system? The in-house team must have the knowledge not only to
ment.” In other words, it is passing on the “recipe” for your prod- recognize any modifications it needs to make to manufacturing
uct, and there is no room for error. For platform processes, plans but also to understand how it might change the regulatory
such as a standard 2,000-liter fed batch, there is not much pathway originally intended for that product.
change in the standard operating procedures (SOPs) and batch
records used from one process to the next. If a process is de-
signed to the platform, and it works at a small scale, a company Summary
mitigates the risks at scale-up. However, for custom processes,
Overall, the decision to stay in-house or to outsource is not
particularly with biologics, it can be much more difficult to main-
easy, but it is quite possibly the most important one you will
tain the integrity of a drug’s formulation. If the integrity is not
make as you begin your journey into the production of biolog-
maintained, that manufacturer suddenly finds itself in a situa-
ics.7 A company venturing into this market might have a promis-
tion downstream where unexpected variables, such as extrane-
ing and potentially life-changing target; yet, it may not have the
ous protein or unpredicted scale-up issues, have a negative
tools to successfully bring it to market. Partnering with a CDMO
impact on the final product.
that offers flexible development and manufacturing solutions
There is much to be said about the advantages of the technical can help reduce the variability biopharma organizations often
knowledge that can be gained from running a variety of campaign encounter during the drug development process; it also creates
types using multiple types of processes, such as in the tech trans- an ability to customize a solution that accounts for the ebb and
fer example above. The experience of analyzing and solving a flow of demand and capacity needs. Rather than relying on a
wide range of process and product issues creates a much stron- strategy that claims to solve forecasting issues, a flexible solu-
ger skill set than only having to address a handful of problems with tion allows a manufacturer to adjust to changes as they occur.
one product type each year. This strategic advantage translates to
If a company determines capacity and expertise gaps exist in its
a higher level of expertise in both operations and development. A
strategy, the advantages of an experienced and flexible CDMO
drug developer must consider how much practice its team has “on
can be invaluable. Having the resources necessary helps miti-
the green,” and if they are prepared with the boldness manufactur-
gate the risks of drug development while creating an opportunity
ing biologics requires. If the decision is to not enter into a partner-
to achieve necessary speed-to-market milestones in a highly
ship, a company is placing itself into a silo and potentially at risk of
competitive market. By accessing pharma’s best weapon against
development uncertainties, a manufacturer reaps the benefits of
As a manufacturer attempts to a cost effective solution that also provides the technical and busi-
ness capabilities needed for commercial success. ■
come up with needed capacity,
the upfront costs of building
infrastructure, hiring personnel,
6 International Conference on Harmonisation of Technical Requirements for
Pharmaceuticals for Human Use, Guidance for Industry Q10 Pharmaceutical
Quality System — https://www.fda. gov/downloads/Drugs/.../Guidances/

and installing the necessary


ucm073517.pdf

7 Lam, Stephen, Managing Demand Uncertainty in Biologics Production

systems begin to mount.


— http://go.patheon.com/rs/910-ISZ-451/images/Whitepaper_FBM_
ManagingDemandUncertainty.pdf

10 www.patheon.com ● doingbusiness@patheon.com
API
Drug
Substance

How Can You Avoid


the Fallout from
Incompatibility
Between Your
API and its
Formulation?

The API characteristics your


formulator can’t ignore

Matthew J. Jones, Ph.D. One difficulty in developing a new drug product is matching the time-
Crystallization Expert line for a drug substance manufacturing development with a drug
product formulation. In most cases, the chemists and engineers will
In drug development, designing a formulation for a drug product (a have almost completed development of a manufacturing process
tablet, for example) calls for careful attention to both the physical when the formulators begin developing the final commercial formu-
and chemical properties of the active pharmaceutical ingredient lation. More often than not, chemists and engineers finish develop-
(API or drug substance). It also requires awareness that certain ment under the assumption that their role is complete — an efficient
process to manufacture the drug substance to a purity specification
physical attributes, such as particle size distribution of the drug sub-
has been developed — leaving the formulator to contend with a drug
stance, can change with processing conditions and changes in the
substance that is less than optimal for product development.
synthesis route that is employed. The formulation, which includes all
of the excipients used to make the tablet, ensures the active ingre- This separation, as well as the lack of overlap in processes, can lead
dient is released in such a way as to not only provide the best effica- to critical knowledge gaps between the chemists and formulators
cy for the patient but also protect the drug substance. If the drug about powder properties. If these drug substance properties are
substance has unfavorable interactions with any of the excipients, it characterized early, it helps to identify and flag any potential issues
can have a negative effect on the shelf life and performance of the that may lead to problems in formulation development. This is be-
drug product and can potentially cause harm to the patient. cause the physical and chemical properties of the drug substance
have a direct impact upon the formulation requirements for the drug
For these reasons, it is critical the experts creating the formulation product. For example, solubility and particle size, both physical
are aware of any reactions that can occur between an API and a properties, control the dissolution rate and therefore the availability
tablet’s excipients. This enables the correct choice of excipients, of the drug substance in the body. Chemical properties can lead to
avoids unpleasant surprises at a later stage and allows the focus to unwanted reactions with other components in the product, resulting
remain upon the manufacturing scale-up of both the drug substance in undesired impurities and loss in efficacy or even harmful side ef-
and the formulated product. So what can a pharmaceutical company fects to the patient. Without an open line of communication between
do to recognize both unfavorable drug substance properties and in- each team, the development chemists are not fully aware of those
compatibility between an API and its formulation? Finding the answer factors that have an impact on formulation development; converse-
to this question early allows a drug manufacturer to avoid potential ly, formulators may not fully appreciate the limits imposed by the
risks to the patient as well as costly interruptions during development. manufacturing process on manipulating drug substance properties.

www.patheon.com ● doingbusiness@patheon.com 11
In addition to a lack of communication, organizational barriers can isolation of the drug substance with the formulation development.
also create challenges. Frequently, chemical development and Doing so can reduce the need for the two teams to go back and
pharmaceutical (product) development are separate organizational forth to optimize the drug substance properties and improve the
units, and good communication is required to minimize the impact of formulation. Yet, many companies struggle to make this transition
this mismatch in the development timelines. due to organizational silos that have become common in the indus-
try. Nonetheless, if a company can remove these perceived walls to
The following list highlights the importance of a drug substance’s communication, it can generate necessary collaboration between
physical and chemical characteristics: two groups whose roles are crucial to successful drug development.

●● Solubility: The solubility of a drug substance in physiolog-


ical fluids — gastric juice or intestinal fluids — is important Communication and collaboration:
as it represents the upper concentration limit for the drug in A solution forward
that environment.
While some may be reluctant to align the processes of API synthe-
●● Dissolution rate: If the material does not dissolve fast sis and formulation because of the perceived risks and costs asso-
enough in the body, it simply passes through with no thera- ciated with front-loading development work, it can be argued that
peutic effect on the patient. On the other hand, if the material the modest cost of gaining good insight into critical formulation-en-
dissolves too quickly, it can become toxic, creating serious abling knowledge early is worth the risk. This is especially true when
safety issues. Achieving optimal absorption of a drug is de- considering that the vast majority of candidates typically fail to make
pendent on the particle properties combined with the formu- it to market. According to a recent BIO Industry Analysis study, “the
lation it is in. overall likelihood of approval (LOA) from Phase I for all developmen-
tal candidates was 9.6% and 11.9% for all indications in oncology.”1
●● Powder properties: Mean particle size and size distribution
Therefore, a company can potentially face added costs even if it
are particularly important because they determine the disso-
does not create a more aligned approach to API synthesis and for-
lution rate, which, in combination with the substance’s solubil-
mulation, and the cost of a failed drug is sometimes too significant
ity, controls the drug’s availability for uptake by the body.
for some companies to overcome.
They also influence the flow properties of a powder.
A simple way to approach this realignment is to create awareness
●● Flow properties: Whether a drug substance is free-flowing, and understanding as well as communication channels between
like coarse table sugar, or has poor flow properties, like con- organizational units. Once these channels are open, it is likely the
fectioner’s sugar, it can have consequences on both the benefits of a more aligned relationship will quickly become appar-
manufacturing process and the product. “Sticky” powders ent. The formulator can now build a substantial base of knowledge
may result in reduced drug substance amounts in the product from the development chemist or engineer during the design of the
due to losses in the processing equipment. Poor flow proper- final isolation process for the drug substance. The formulator will
ties of the drug substance-excipient mixtures might lead to also become more aware of what factors have an impact on formu-
segregation of the components and insufficient content uni- lation development. At the same time, the development chemist will
formity in the individual dose. have a deeper understanding of the chemical properties of the API,
which helps identify possible chemical interactions with the excipi-
While it may seem like, in the case of incompatibility of physical at-
ents the formulator intends to use. The alignment ends up becoming
tributes, the easiest solution is to redevelop the final isolation to fit
a small step toward implementing procedures that can be critical to
the formulation, extensive changes to a process would likely require
the overall process. As a result, this communication of information
additional clinical studies to demonstrate equivalence of the product becomes a key to identifying potential issues and working together
for approval. Even minor modifications are not feasible once the API to resolve them.
manufacturing process has been locked and even registered with
the regulatory authorities. For a small company pursuing a new Implementing significant changes such as these to a company’s
drug, there may not be funding for additional clinical trials. This procedures can be difficult, and without the money and expertise,
could also cause considerable delays in the development timeline, it can even be impossible. An alternative is to seek a partner that
which is something pharmaceutical companies often cannot afford. has already considered and addressed this issue. Working with an
experienced partner obviates the need for time-consuming and po-
With a typical time to market of 12 years, any delay reduces the al- tentially disruptive organizational changes. It also allows rapid gain
ready short time for recovery of investment once the product is com- of understanding and time by making full use of your partner’s
mercialized. Decreasing development timelines by generating the knowledge and experience when defining the best strategy for a
right type of information early in the development process not only seamless development from drug substance to drug product. ■
expedites a company’s ROI, but also, and more importantly, gets
the medicine to the patients faster. One measure to prevent the 1 Biotechnology Innovation Organization (BIO), Clinical Development Success
Rates 2006-2015 — https://www.bio.org/sites/default/files/Clinical%20
fallout of incompatibility or unsuitable API properties, and thereby Development%20Success%20Rates%202006-2015%20-%20BIO,%20
timeline delays, is to align the development of the synthesis and final Biomedtracker,%20Amplion%202016.pdf

12 www.patheon.com ● doingbusiness@patheon.com
API
Drug
Substance

Trends and
Challenges in
Outsourced
Oral Solid
Dosage Forms:
A Q&A

Anil Kane, Ph.D., MBA


Global Head of Technical and Scientific Affairs,
Formulation

Combination drugs, ingestible sensors, and targeted deliv- clinical benefits are also under evaluation. And, pediatric-friend-
ery are the future of pharma. ly formats such as sprinkles, mini-tablets, and orally disintegrat-
ing tablets are gaining popularity.
As drug companies have fewer and fewer new compounds
entering their R&D pipelines, outsourcing of development In addition, the demand for lifecycle management strategies
and manufacturing activities for oral solid dosage forms and such as pediatrics, controlled-release dosage forms, fixed-
sterile forms is on the rise. In a recent podcast, Anil Kane, dose combinations, and formats such as multi-layer tablets or
Ph.D., MBA, Global Head of Technical and Scientific Affairs multiparticulate capsules is increasing.
of Formulation at Patheon, discussed trends and significant
changes in outsourcing the manufacture and development Due to more stringent regulatory requirements, developers
of solid oral dosage forms. need to apply a much higher level of product and process un-
derstanding and robustness justification by using systematic
scientific- and risk-based approaches as well as the principles
Q: What have been the most significant
of quality by design (QbD).
changes in outsourced oral solid dosage
forms over the past decade? What are the Additionally, increased use of highly potent compounds has led
current trends? to the need for full containment, process automation, closed-
loop product transfers between processes, barrier isolators,
Kane: The shrinking new drug discovery pipeline has led to and equipment with clean-in-place and wash-in-place capabili-
significant changes in dosage form development and manufac- ties. Moreover, early-stage investment in and adoption of con-
turing strategies. Existing molecules are being evaluated for tinuous manufacturing is gaining popularity. Finally, there is a
newer indications in the same or different therapeutic catego- need to address data integrity requirements, resulting in more
ries. Fixed-dose combinations of new drug entities combined automation, data capture, documentation control, and training
with off-patent drug candidates for synergistic effects or other to imbibe the culture of quality.

www.patheon.com ● doingbusiness@patheon.com 13
Q: Can you highlight any changes you have The industry will
seen in lifecycle management strategies?
slowly embrace the
Kane: Some of the most widely used lifecycle manage-
ment strategies are pediatric, controlled-release dosage long-term benefits of
forms, and fixed-dose combinations. Pediatric formulations,
particularly in terms of palatable powder for reconstitution, continuous manufacturing
sprinkles, dissolvable tablets or mini-tablets, and chewable
soft gels, are becoming increasingly popular.
and will invest in this model
Controlled-release technologies are a frequently applied
to develop clinical and
lifecycle management extension strategy. These technolo-
gies have apparent benefits in delivering drugs over an ex-
commercial products.
tended period of time, reducing the frequency of dosing and
improving patient compliance. Another lifecycle manage-
ment strategy is solubility enhancement, thereby potentially to Phase III, but these programs have continued at Patheon
lowering the dose of existing drugs. and we continue supporting these programs from clinical
through registration and then commercial launch, and resup-
ply. This ensures continuity of supply, as well as avoids loss
Q: What trends has Patheon seen in of know-how and product knowledge.
controlled-release dosage form development?

Kane: In the last three to five years, we have experienced Q: What are future trends you expect in the
an increase in the demand for dosage forms that exhibit an pharma industry?
enteric release followed by an immediate-release portion
and then a slow release of the drug over a specified period Kane: I believe there will be more emphasis on continuous
of time. These delivery systems could be for the treatment of manufacturing of solid oral dosage forms. The industry will
irritable bowel syndrome, ulcerative colitis, Crohn’s disease, slowly embrace the long-term benefits of continuous manu-
or certain types of cancers in the lower part of the GI tract. facturing and will invest in this model to develop clinical and
commercial products.
The benefits of these oral drug delivery systems are that drugs
can act locally as well as systemically by getting partially or ●● There will be emphasis on patient-friendly dosage forms,
completely absorbed at the site of action, thus improving ther- reducing pill burden, addressing the problem of polyphar-
apeutic effect. Dosage forms that have been formulated to macy, and improving patient compliance.
deliver such drug candidates include enteric-coated con-
trolled-release tablets, or pulsatile-release beads in the form ●● Combinations of drugs and devices will bring several
of extruded beads, pellets, and capsules. benefits to patients.

●● Drugs targeted to specific sites of activity or absorption


Q: What is the effect of mergers, will enhance the therapeutic effect use of biodegradable
sensors, imaging techniques, and application of medical
acquisitions, and licensing deals on an and pharmaceutical electronics to deliver drugs accurate-
outsource partner? ly and at a pre-determined rate for enhanced efficacy.

Kane: The mergers and acquisitions as well as in- and out- ●● Ingestible sensors could monitor the compliance of drug
licensing activity in the pharmaceutical industry continue to administration and drug misuse.
grow. As more clinical candidates are outsourced, Patheon
has seen that clinical candidates and the drug products are ●● Technological advances can help personalized therapy
licensed out by small or emerging companies to large com- and delivery of the right drugs for improving therapy. ■
panies or biotech companies, and then get acquired by other
pharma companies. But, the programs stay with us despite
change of ownership. In the past three to five years, we have
seen more than 55 programs licensed out between Phase I

14 www.patheon.com ● doingbusiness@patheon.com
Formulation
Development

Characterizing
DS Properties
Early Can
Optimize DP
Formulation

Anil Kane, Ph.D., MBA


Global Head of Technical and Scientific Affairs,
Formulation

Early on, the drug substance (DS) manufacturer will provide a Siloed development is neither
wealth of preliminary characterization data. And if the drug is
efficient nor effective
active in early nonclinical and preclinical testing, a larger quantity
of the DS will be prepared for more comprehensive animal safety Traditionally, DS and DP have related but distinct deliverables. The
and pharmacokinetics studies. These studies may require up to end-product for a DS development team is a pure chemical sub-
several hundreds of grams (sometimes a kilo) of purified DS with stance produced in 5-to-12 synthetic steps; the end-product for a
low levels of impurities and residual solvents that meet global DP development team is a patient-friendly drug. For DP formula-
regulatory standards. tors, the DS team’s end-product is their starting point.

But changes in the DS process as it scales up can affect the drug Typically, the industry has been conservative in characterizing the
product (DP). As processes change, many properties of the DS full properties of the starting material for the DS: A minimum num-
can also change: its purity, potency, by-products, particle size ber of tests are conducted to suit the requirement of the immediate
distribution, morphology (crystal shape and structure), and rate next step, such as preclinical toxicity or animal exposure studies.
and extent of dissolution. Therefore, as DS manufacturers evalu-
ate and optimize the synthetic route, process conditions, crystal- This is unfortunate. As molecules become more difficult to formu-
lization solvents, etc., they must understand and track these late, this sequential, compartmentalized development model is no
changes, and discuss them with DP formulators to anticipate longer adequate.
challenges in formulation.

DS characterization is critical to DP formulation but characteriza- DS and DP functions must collaborate on a


tion and formulation are often not integrated during drug devel- sound formulation strategy
opment. This creates needless difficulties because if DS chem-
ists and DP formulators collaborate on a formulation development Early stage, pure DS is ideally suited for characterization tests
strategy for early Phase I first in-human clinical studies, they can which can catch problems early by identifying properties that could
save time, money, and avoid rework. dictate future formulation choices. Yet developers routinely assume

www.patheon.com ● doingbusiness@patheon.com 15
the preclinical period is too soon to consider formulation, reasoning know whether the molecule is stable at the pH ranges prevail-
that “We are not formulating yet; we are just identifying challenges ing at its intended site of release, absorption, or permeation.
before we trip over them.” A formulator can use this information to modify the pH of the
microenvironment to improve solubility of the DS, or prevent
A systematic DS characterization should include a series of critical precipitation in biological fluids of certain pH ranges.
formulation-enabling tests, and establish a thorough understand-
ing of a molecule’s properties to flag issues that can be addressed ●● pH Stability: The stability of a DS under various pH conditions
early in the clinical stages. A detailed DS profile can help DP formu- is as important as its solubility. Formulators can add acidifiers or
lators craft a strategy to make sure, for example, that the DS will not alkalizers to stabilize the DS in the relevant dosage form.
get degraded in the GI tract. A forced degradation study can ensure
a molecule is protected via its formulation. ●● Partition Coefficient (log P, log D): This tells the formulator if
the molecule will dissolve better in a lipid or an emulsion. For
For example, recently a company produced a DP formulation in par- poorly soluble lipophilic compounds, formulators can develop a
allel with the human Phase I clinical study, based on a truncated DS lipid-based formulation, using surfactant, self-emulsifying sys-
characterization effort (shortened to reduce overall development tems, or other techniques. If a compound has better solubility in
time). The resulting formulation was a DS blend in a capsule, packed lipids than it does in aqueous buffered solutions, formulators
in blisters for stability. However, after one-month stability data can perform a systematic lipid vehicle screening test using
showed that the DP had degraded, a thorough investigation and root phase diagrams to understand its solubility in mono, binary, or
cause analysis revealed that the DS was degrading due to alkaline tertiary solvent mixtures, and then formulate a dosage form bet-
conditions, and in response to exposure to a specific range of light. ter suited to deliver the drug without precipitation. A systematic
lipid digestion screen can ensure solubilization/emulsification of
Due to the rushed timeline and incomplete characterization effort, the drug in biological fluids on administration.
there were no baseline characterization data available that demon-
strated the stability of the DS in different pH conditions, or in the ●● Ionization Constant (pKa): The pKa of a drug influences many
presence of different intensities of light. This was an avoidable mis- biopharmaceutical characteristics such as lipophilicity, solubility,
take that required reformulating the DP and slowed development. protein binding, and permeability. These in turn directly affect
Systematic testing could have prevented it. pharmacokinetic characteristics such as absorption, distribution,
metabolism, and excretion. For instance, a molecule can be
Data from forced degradation studies enable chemists and formula- modified to link to a certain protein, such as complexation, or
tors to understand if a DS is stable under acidic or alkaline pH buf- salts such as chloride or maleate. Knowing the pKa is important
fers, as well as in accelerated conditions of light, heat, humidity, and for salt screening, an early step in determining which candidate
exposure to oxygen. If a systematic forced degradation study on the to advance (or terminate). And it helps formulators optimize drug
DS and DP had been carried out early on, the formulators could delivery. Different salts of a free base may have different solubili-
have designed the DP with excipients to stabilize the DS in an alka- zation, absorption, and stabilization properties. A systematic salt
line environment. And if a DS were sensitive to light, it could be for- screening of a DS in early stage is key to identifying the right salt
mulated in a capsule with light protection, or in a tablet with an to use to take development to the next steps.
opaque coating. It may need to be manufactured under specific light-
ing conditions. For example, a recent case study of a DS revealed
that it was stable under red light but not yellow. As a result, handling
the DS in the lab, as well as during manufacturing, was only done
under red light to maintain DS stability in the DP, and to avoid any
degradation and the formation of photo-catalyzed impurities.

Here are some crucial DS properties that can inform and improve
formulation strategies, but only if the testing is done in a timely
fashion and results are communicated.

Properties critical to formulation that are


not often available early (but should be)
●● Aqueous And pH Solubility: Solubility of the material in
acidic, neutral, or alkaline pH buffers helps DP formulators

16 www.patheon.com ● doingbusiness@patheon.com
Other physico-chemical properties ●● Compressibility: It is useful to understand the behavior of the
DS under compression. For example, do the particles fracture?
critical to formulation
Do they deform? Is the material ductile, fragile, and does the
●● Particle Size And Morphology: The size and shape of parti- blend have sufficient elastic or plastic properties? Based on the
cles determines their surface area and density, which in turns answers to these questions, a formulator can add suitable excipi-
influences the flow of powders. While the shape of many crys- ents such as cellulose derivatives, dicalcium phosphate, and/or
talline substances changes due to the conditions of crystalliza- various grades of lactose or mannitol to optimize the compress-
tion, crystal size is primarily a function of the rate of cooling. ible behavior of the finished blend for a tablet formulation.
Limiting size to a narrow range can ensure uniform blending
with excipients in the formulation. Particle size also affects sol-
ubility; micronization is a common first tool to apply to Properties that DS chemists routinely
poorly-soluble substances. The dose of the drug and the drug provide (and are not likely to change)
loading both impact formulation. For example, a low-dose drug
or a highly potent drug administered at a very low concentration ●● Melting Point: A low melting point substance may need to
are both examples of drug candidates at high risk for problems be handled differently by the formulators during dosage
in blend uniformity, “content uniformity,” and segregation/ form development.
non-uniformity in the finished product. A high-dose formulation
or a high loading dose formulation present fewer challenges in ●● Organic Solvent Solubility: Important to understand for
terms of blend uniformity, but increase the risks of product pro- specific cases such as if the compound must be spray dried
cessing and compressibility problems. Particle size distribution by dissolving it in an organic solvent, or if a solvent-based
becomes more important as the DS is formulated into the DP. drug layering or polymer coating is to be applied.

●● Stability Under Forced Degradation: The stability of the


DS under accelerated conditions of acidity, alkalinity, light, DS chemists and DP formulators must
humidity, and temperature is important to formulation of the communicate early and often
dosage form. Knowing it helps the formulator select the right
ingredients and excipients, as well as the process conditions To succeed in an increasingly complex environment, companies
that will ensure stability of the product during manufacture. need a more holistic approach to crafting their formulation develop-
ment strategy. Ideally, DS chemists investigate properties in paral-
●● Long-term Stability: The stability of the DS under ICH condi- lel with DP formulators, and characterize things that will not change
tions over a longer period also helps formulators select excip- (e.g., log P, log D, solubility, MP, pKa, etc.). Meanwhile, DP formula-
ients and process conditions that maintain the stability of the tors determine the best solution for formulation, and tell the DS
product during manufacture, and in subsequent storage. chemists what properties they are looking for, such as direct com-
●● Hygroscopicity And Water Sorption: If the drug substance ab- pression, limits on particle size distribution, and polymorphs.
sorbs moisture under ambient and plant conditions, it will need to
As development proceeds, strong lines of clear and frequent com-
be protected from normal humidity. It is also important to know the
degree of moisture pick up, and if it is prone to convert to hydrates, munication can screen out risks because DS chemists can inform
solvates, or to other polymorphs. Different solvates, hydrates, or DP formulators if something critical has changed. The two disci-
polymorphs of the same parent drug can have different solubility, plines also can collaborate by agreeing on high-risk areas, commit-
rate of absorption, or stability profile characteristics. A dynamic ting to a sound set of characterization tests, and bracketing the DP
vapor sorption (DVS) study can help understand the molecule’s characteristics needed or preferred.
sorption and desorption properties under various humidity levels.
Molecules continue to get more challenging from the perspective of
●● Flow properties: Bulk and tapped density are important for for- solubility, bioavailability, and exposure. Advances in formulation
mulators because they indicate the flow of powder into the dies
technologies continue to provide more solutions. Necessity is the
of a high-speed tablets press, the potential of the blend to seg-
regate, and to have compressibility challenges. When a DS ex- mother of invention, but recent inventions have drawn companies
hibits poor flow characteristics, formulators can select excipients into developing more difficult molecules.
that improve flowability (e.g., Fast Flo lactose/MCC), or reduce
the inter-particulate forces with colloidal silica, among other sub- It’s time to establish effective communications between DS
stances. They can select process steps that adjust the particle chemists and DP formulators if we are to manage the risks and
size of the blend, such as wet or dry granulation. Flow properties maximize the rewards of transforming less soluble and less bio-
become more important as you scale-up manufacturing. available molecules into effective new drugs. ■

www.patheon.com ● doingbusiness@patheon.com 17
Pharmaceutical
Process
Development

Avoid the Do-Over:


Why Early
Investment
in a Scalable
Manufacturing
Process is Critical
Title Goes Here

Enrico Corona Jonathan Sutch


Director, Senior Manager,
Pharmaceutics & Process Technology Formulation Development

The road to take a drug compound from discovery to commer- The problem with shortcuts
cialization is long, expensive and often fraught with unforeseen
challenges. While every project will undoubtedly face some Small companies often hesitate to invest in a full analysis of
bumps along its path, far too many programs hit insurmount- their molecule’s physical and chemical properties in early stag-
able obstacles that require innovators to backtrack and correct es for a variety of reasons. Unfortunately, this delay can create
their course before proceeding, further extending timelines and roadblocks as the compound matures.
adding costs.
Most small biopharmaceutical companies, for instance, function
The failure of drug compounds in mid- to late-stage develop- with accelerated development timelines and limited resources.
ment is far more common than anyone would like it to be. Only This environment pushes them to race to the next development
about 30% of drug compounds successfully transition from milestone that’s linked to funding. In addition, many biopharma
Phase II to Phase III, and just 54% of the remaining programs companies simply may not have enough API available in Phases I
ever advance beyond Phase III.1,2 Reasons for Phase III failure and IIa to run all the necessary process characterization studies.
span the gamut from safety challenges to poor efficacy to com- Companies are often working on life-saving treatments, generics
mercial complications. In addition, some firms learn late in the or biosimilars, which adds great pressure to advance molecules
process — during Phase III or IV, when they should be focused to market as quickly as possible.
on evaluating drug safety and efficacy — that fundamental
problems in process design must also be addressed, resulting In the end, smaller companies often trade investment in a prop-
in a repeat of drug product development activities.

How can developers avoid these costly delays? Investing in a 1 J. Paul, “Reducing the Risk of Late-Phase Failures,” Pharma R&D, July 5,
2016, https://pharma.elsevier.com/ pharma-rd/reducing-the-risk-of-late-
scalable manufacturing process early-on can make the differ- phase-failures.
ence between advancing through drug development in a pro- 2 T.J. Hwang et al., “Failure of Investigational Drugs in Late-Stage Clinical
ductive, step-wise fashion and having to redo early-phase work Development and Publication of Trial Results,” JAMA Intern Med.
176(12):1826-1833 (2016), http://jamanetwork.com/journals/
to correct problems. jamainternalmedicine/article-abstract/2565686.

18 www.patheon.com ● doingbusiness@patheon.com
erly characterized molecule for a “quick and dirty” approach to
their API. We’ve seen such firms use very conservative pro-
It is critical to ensure that
cess parameters during development, which generates a limit- process development is robust
ed pool of data. Such narrow process parameters may provide
fast answers that scratch the surface of process information, and manufacturing scale-up is
but will not collect the richer data that is needed for adequate
characterization and prediction of success during scale-up. embedded in from the
A major problem with this strategy is that drug developers ab- beginning ... [and] weigh the
solutely need a great deal of information about their molecule
for it to be successful in later stage work. Indeed, the key com-
short-term benefits against the
ponent in any drug product is the drug substance. Not under- potential long-term risks —
standing its stability, crystallinity, solubility, hygroscopicity and
other parameters will only spell trouble during process scale-up especially to patients.
down the road.

To put this into perspective, a company that takes what may


be its first or second batch of material into development could
known example of conformational polymorphism from the
very well be working with an unfavorable polymorph or salt
mid-1990s. The API changed polymorph while it was in com-
form that stands no chance of clinical (or commercial) suc-
mercial production and the life-saving HIV treatment was
cess down the road.
forced to be withdrawn from the market until developers
In addition, not thinking about a molecule’s future from a pro- could solve the problem.
cess design standpoint means that the next batch created
Drug makers must learn lessons from situations like these;
could have a completely different set of characteristics from the
when they’re developing products, it is critical to ensure that
one already in development. Such characteristics may include
process development is robust and manufacturing scale-up is
a different particle size or shape, solubility variations, or even
embedded in from the beginning. Any firm considering the path
different polymorphic forms, which undoubtedly will affect drug
of delayed process development work should strongly weigh
safety and efficacy.
the short-term benefits against the potential long-term risks —
The only way to move forward at this point is to spend time especially to patients.
and money to redevelop the formulation. While very ear-
ly-phase redevelopment is not a problem, if companies are Phase III: Still work to be done
moving into later development without having done proper
characterization studies, they essentially have the wrong drug Big Pharma companies, with reduced budget constraints, usu-
in their formulation. ally allocate resources to performing sufficient process charac-
terization studies that allow for better process design beginning
Such a lack of preparedness could pose a variety of potential in early-stage work. Some drug developers even use a platform
issues and consequences. We’ve had Phase II projects trans- approach that allows them to standardize solubility, compound-
ferred into Patheon with the hopes of starting Phase III produc- ing, lyophilization and various critical parameters across a
tion immediately. Sometimes, at this point, even a small but range of compounds.
necessary change requires us to go back and conduct rounds
of formulation work, which delays the timeline by several Even if such groundwork has been laid by an in-house team, it
months and costs of hundreds of thousands of dollars. If a for- is still possible for an experienced partner to determine further
mulation vulnerability were to be identified at an even later areas for improvement through risk assessments during late-
stage, it would cost much more to rectify. stage tech transfer to a CDMO. We feel it is important during
this critical juncture to ensure a compound will both be suc-
Even products already on the market are not safe from po- cessful as a commercial product and has the necessary data to
tential manufacturing problems if the process is not robust satisfy the regulators — another area where time can be lost.
enough to handle common alterations like a change in the
supply chain. When changes require a product to come off This is where the principles of quality by design (QbD) really
the market, it will cause a significant loss in revenue and come into play. Regulators are now expecting to see an en-
consumer confidence. Ritonavir (Norvir, Abbott) is a well- hanced level of product knowledge to ensure robustness —

www.patheon.com ● doingbusiness@patheon.com 19
and Phase III is a good time to do that with the help of a manu-
facturing partner.

If you perform QbD work and evaluation during Phase III, then
chemistry, manufacturing and controls work will not be a
rate-limiting step when the clinical program is ready to seek
regulatory approval.

Real-world failures and successes


Compounds and process design cannot be created for the mo-
ment; they must be developed with the future in mind. When
firms develop a compound — no matter how early-stage the
project may be — they must do so with an eye on the informa- more robust as it scales up and better able to handle changes
tion required for successful commercialization. such as those in supply. Engaging QbD approaches — such
as the use of risk assessments, experiments of design, and a
The following real-world scenarios demonstrate how the choice good control strategy — early in the process will help ensure
to invest (or not to invest) in a scalability strategy in early devel- that all the stages of scale-up can be managed throughout a
opment has significant long-term effects on the project. product’s lifecycle.

Choosing speed over process development: Waiting to understand key product


Repeating early-stage work. attributes: Costly reformulation.
Close to commercialization, Company A transferred a product During a transfer, we learned that Company B was working with
to Patheon while simultaneously switching their drug sub- a formulation it already knew had dissolution challenges. The
stance provider. This supplier change had huge repercus- client felt it was not worth investigating the problem until later
sions on the API that the developer was unable to foresee phases to avoid development delays. We helped the company
because of its very light early development work. Rather than trace the problem back to a fundamental issue with their formu-
starting late-stage production as the client had hoped, lation. If left unaddressed, the product would not have passed
Patheon had to perform re-formulation work to better under- regulatory scrutiny, so the only path forward was reformulation.
stand the API and make sure the formulation was more robust If Company B had not ignored the dissolution issues and invest-
before moving forward. ed earlier in the right level of science, this backtracking could
have been avoided.
This delay could have been avoided by applying the principles
of QbD to the drug product and the drug substance. A product
that is developed using QbD principles is naturally going to be Implementing QbD with FMEA:
Successful scale-up.
As Company C moved through the development process, QbD

When firms develop a was an important priority. Not only does a QbD-type submission
give regulators confidence in the submission’s ability to handle fu-

compound — no matter how ture changes, but it also gives manufacturers confidence that their
process is robust. Robustness is a central part of successful
early-stage the project may scale-up and using appropriate QbD strategies enabled Company
C to fully understand its product as it scaled through development.
be — they must do so with One particularly effective tool in the QbD toolbox is failure mode
an eye on the information and effects analysis (FMEA), the gold standard in risk assess-
ments for understanding any possible design failures. A well-exe-
required for successful cuted FMEA approach should be interdisciplinary, tapping into the
knowledge of experts from development and commercial teams.
commercialization. Checkpoints of assessment during formulation development may

20 www.patheon.com ● doingbusiness@patheon.com
involve numerous process parameters and the effects they could
have on clinical quality attributes like safety, quality and efficacy.
Step-wise and gated
Step-wise and gated assessments help companies build quality
and science into process design; they have broad knowledge of
assessments help
how API changes may affect quality attributes, for instance, or if a companies build quality
change in excipient grade will affect the end-product.
and science into
Given all the areas involved in an effective FMEA, working with a
knowledgeable CDMO can be advantageous. Remember, a process design.
small company may have access to just three or four molecules
over the course of several years, but a CDMO like Patheon will
The single-source option enables the CDMO’s drug product
see hundreds of different molecules in the same period. This
team to provide feedback to its drug substance team about
makes us adept at handling challenges, knowing which potential
what worked well and what did not with the API. Company E, for
problem areas to look out for during risk assessments and lever-
instance, might benefit from addressing some powder flow is-
aging more diverse information for better process development. sues at the Phase II juncture and still ultimately achieve the
desired end-point (i.e., high yield and a pure drug substance)
along with deep knowledge about key product attributes. Con-
Failure to consider commercial equipment:
trast this situation to an API delivered from a third party. If flow-
Development delays. ability problems surfaced in this scenario, it would take much
more time to investigate the source of the issue and adjust the
Company D used its in-house team to develop a lyophilized
process design appropriately. Without this kind of network, a
formula. The firm’s long lyophilization cycle was fit for purpose
CDMO might simply just take whatever drug substance it re-
at the early stage, but became economically unreasonable as
ceives and work with that material — one that may have red
the process was scaled up. Additional development work was
flags it would not find without a deeper investigation.
needed to rethink the lyophilization process.
In the end, being able to influence the type of drug substance
The firm missed out on the fact that having full knowledge of and drug product through a single network of experts can
equipment trains — and the capability of full scalability between make the development and scale-up path much more effi-
laboratory and production lyophilization units — would lead to cient and, with deeper knowledge, a more robust process
better development of freeze-drying cycle parameters as well will be developed.
as cost savings. By understanding the full breadth of equipment
required from batch through commercialization in early stages,
companies may spend a little more time developing the right Summary
process at small-scale, but scale-up will be far easier later on
In an effort to move through the drug process quickly, some
with potentially reduced cycle times.
companies spend less time doing proper development work.
This scenario applies beyond lyophilization as well. We’ve seen The compound may arrive at the tech transfer commercializa-
first-hand that deep knowledge of various equipment trains and tion scale with a dossier that appears scientifically sound. But
the different parts that connect them — as well as access to a when the system is put under a stress it has not encountered
full breadth of tools and techniques, starting from benchtop to before, the process may experience significant problems if it is
not robust. Patheon believes this situation can be avoided by
clinical manufacturing and up to commercial manufacturing —
partnering with an experienced development team and ad-
will greatly contribute to the success of nearly all molecules.
dressing scalability and process design early in development,
and building on this work through to commercialization. This
Using a single-source vendor through investment will pay dividends as one takes a compound into
late-stage commercialization with a good understanding of the
scale-up: Time and cost savings.
compound and process. ■
Company E had developed its first GMP batch of API and success-
fully used it for the first batch of clinical supply. The firm, ready to
begin Phase II trials, had an opportunity to build in additional under-
standing by working with a CDMO provider that aligns drug substance
and drug product units (such as the Patheon OneSource™ program).

www.patheon.com ● doingbusiness@patheon.com 21
Pharmaceutical
Process
Development

Solving the
OOS Problem
with
Continuous
Manufacturing

Eric Jayjock
Director,
Continuous Manufacturing

The goal of Pharmaceutical Process design has long been to tal, equipment or material-based, is practically never investigat-
implement a fixed process that can produce on-spec materials, ed. To understand the impact of those on the manufacturing pro-
which can be confirmed by quality testing. cess, and the challenge they present, it is important to have a
clear view of how a product moves through its lifecycle.
However, there is variability in everything. In the manufacturing
world, variability can be produced by environmental conditions The standard mode of drug manufacture is to begin by produc-
(such as seasonal changes), the way a particular piece of ing batches in the 1kg to 10kg range, and then scale up by a
equipment performs through its lifecycle, or even the properties factor of 10 for pilot scale experimentation, and another factor
of the incoming materials (their density, or flowability). Conse- of 10 for commercial scale production, which typically ends up
quently, the combination of a fixed manufacturing process and somewhere between 150kg to 800kg.
normal variations in environment, equipment, and material
At each step up the scale, batches are produced over a relative-
properties can, and often do, produce products that will fall out
ly short period (when compared to the full production lifecycle),
of spec (OOS).
often with API material that is still undergoing process changes.
Fortunately, the data-rich nature of the continuous manufactur-
When the manufacturing process is scaled for commercial pro-
ing process potentially can greatly reduce or even eliminate
duction of 150kg to 800kg batches, there is very little chance to
OOS events.
understand the impact of the variability introduced by incoming
materials; it is both logistically difficult and cost prohibitive to
The complexity of the product lifecycle interrogate the process with batches of that size. Once the pro-
cess has been validated, it is essentially fixed, making it difficult
Strategies exist for managing the variables introduced by envi- to determine why batch failures happen. And in those cases
ronmental factors, and by equipment, but they usually do not ex- where the cause of a failure can be identified, it is often prohib-
tend to dealing with variations in material properties. Moreover, itively complicated to roll the accumulated product knowledge
the interactions between normal variations, be they environmen- back into the manufacturing process.

22 www.patheon.com ● doingbusiness@patheon.com
That is why OOS is an inevitable outcome of a fixed manufac- Here’s how it would integrate into a process development study:
turing process. Any variation in material properties not investi-
gated during development can lead to batches of OOS material, 1. Perform a risk analysis (FMEA or a similar method) for the
with its attendant waste and costs. product under investigation using any and all available
date to estimate the intended process knowledge space.

Two reasons why continuous 2. Use one run of about 300kg where a different set of ex-
perimental conditions is established once every 10 min-
manufacturing makes every kind of sense
utes or so (this time will be dependent on the amount of
Continuous manufacturing for oral solid-dose (OSD) products back mixing within the process).
in particular (and pharmaceutical manufacturing in general) can
make manufacturing more reliable, and less wasteful and cost- 3. Assimilate the results of changing about 30 conditions to
ly than the batch method for two reasons: get the big picture of how they act individually, or in com-
bination to affect the output.
1. Continuous manufacturing de-couples batch size from
the amount of material needed to examine the integrity of 4. From that, refine the knowledge space and define a control
the process, allowing commercial production to be stud- strategy that will keep the process within a design space.
ied in more depth with much less material. This control strategy may involve closed loop control (of the
entire process) if a particular anticipated variation has a
2. Steady-state unit operations (feeding, blending, compac- significant impact on a critical product quality attribute.
tion, and so on) make it much simpler to characterize a
process as it does not vary over time. This makes it easier 5. After production starts, continue to map process parame-
to relate material attributes to process conditions and ters to product properties. When those relationships
product attributes. change, assess the quality of the product, and investigate
the possible causes of change, including material varia-
The consequence of these two effects is that quality by design tion (that is, strive to develop a more nuanced under-
(QbD) process understanding can be built on a scale more typ- standing of the process).
ically associated with pilot scale quantities than with manufac-
turing-scale production, with a concomitantly lower investment 6. If necessary, integrate new knowledge into the control
in time and money. strategy going forward.

Six steps to implementing The big benefits


continuous manufacturing Money is always an issue in pharmaceutical manufacturing,
The conventional approach to pharmaceutical manufacturing is and continuous manufacturing can save lots of it. On one cur-
largely a trial-and-error process that identifies an acceptable rent project, continuous manufacturing reportedly saved tens of
range of process parameters that will deliver the expected millions as the company did not have to run multiple manufac-
product attributes. Continuous manufacturing allows one to in- turing-scale development batches during process develop-
vestigate product and process performance, incorporating from ment, thereby reducing material costs.
three to seven factors (with 20 to 50 conditions), using about as
But perhaps more important than cost savings, continuous
much product as a single conventional batch produced by the
manufacturing can gather knowledge at every stage of a prod-
conventional process.
uct’s lifecycle (from development through production) and loop
For example, with a typical direct compaction tableting set- it back into a single, risk-based control strategy. That leads to a
up, the transit time on the line might be a little less than general improvement in quality, and, consequently, fewer rejec-
10 minutes, at a throughput around 50kg an hour. With a tions. The latter is particularly important because the majority
continuous solid-dose manufacturing process — integrating of failed batch investigations end up with no root cause analy-
feeders, mills, blenders, and a tablet press — it is possible to sis, according to the FDA’s Center for Drug Evaluation and Re-
change one or more aspects of the process line, wait until search. Often, when manufacturing conditions change enough
the time for the transition, and then collect a sample. In ef- to throw a product out of spec, the manufacturer never finds out
fect, 30 independent experiments can be performed with what specific conditions have changed. That is hardly a recipe
about 300kg of product. for improvement, and it is a leading cause of batch failures.

www.patheon.com ● doingbusiness@patheon.com 23
more intricate than that employed in the traditional batch model,
so cleaning is both more difficult and more time-consuming.
However, the time gains that come with continuous manufactur-
ing more than make up for that.

Patheon’s commitment to the future


Patheon is designing and building continuous manufacturing lines
today, incorporating several features to overcome those barriers.

We are designing the lines in a modular fashion, allowing equip-


ment (such as feeders and blenders) to be switched out as de-
By continuously improving the process, a manufacturer can re- sired. This makes changeovers faster as clean units can be put
duce end-product testing — or even eliminate it over time — as the on the line while the ones they are replacing are being cleaned.
process becomes increasingly well understood, and controlled. A modular design is also more flexible, allowing the process
flow chart to be customized for each product. If a product works
well with a particular make and model unit — or if the product
Well, if it’s all so easy… was validated on that unit — then it can be installed easily in
any line making that product.
Actually, it’s not so easy, and the barriers to adopting and imple-
menting continuous manufacturing are varied. Patheon is also taking responsibility for the design and devel-
opment of the supervisory control system responsible for
Inertia leads the list of challenges. Pharmaceutical manufactur- coordinating unit operations. Again, because the process is an
ing processes and techniques have changed little over the last integrated one, these responsibilities can no longer be left to
50 years. Part of the reason for that is that the industry is highly individual equipment vendors, and Patheon has worked to build
(and appropriately) regulated, and regulatory agencies this capability internally.
traditionally approved process applications based on previously
approved approaches. Today, however, the FDA has begun Also, we are building up our ability to do risk-based process and
championing emerging technologies for pharmaceutical manu- product development for OSDs. Because Patheon possesses
facturing, and right now there are two FDA-approved continuous this knowledge internally, companies will be able to continue to
lines in operation: Vertex, which received approval for the cystic update and improve processes to ensure the highest level of
fibrosis drug Orkambi in 2015, and Janssen, which in April 2016 quality, even after filing, secure in the knowledge that the
received FDA approval to move its drug for HIV-1 drug infection changes they make will not adversely affect production or prod-
from batch to continuous manufacturing. The European Medi- uct quality. Indeed, Patheon is aiming to implement dynamic
cines Agency (EMEA) has been similarly supportive and has ap- processes that can adjust to keep products in-spec in the face
proved the same Vertex process for Orkambi. of any variation, including natural ones.

Inertia also stems from long-held cultural beliefs and norms. In the scientific community, we tend to establish ideal standards
The pharmaceutical industry has been accustomed to look to and assume that, with enough attention to detail, we can always
its vendors for equipment expertise, even as it expects its de- produce to them. However, perfection is not attainable. In phar-
velopment scientists to optimize each unit’s operation. Continu- maceutical manufacturing, there will always be variations in in-
ous manufacturing, however, is an integrated process and must gredients, or changes in temperature and humidity, that can
be designed and run holistically. In fact, just like nearly all other work to frustrate the best-laid plans, not to mention scientific
manufacturing industries (oil, chemical, food, agriculture, and ideals. With continuous manufacturing, we can monitor product
so on), continuous manufacturing ultimately demands a leading quality continuously, and make adjustments in real time to keep
role for Process Engineering. the product in spec. And when it isn’t, as will sometimes (albeit
rarely) happen, we can step in more quickly than ever before to
Accordingly, to reap the benefits of continuous manufacturing, limit the volume of OOS product — saving time, money, and
the pharmaceutical industry will have to assume responsibility staying ever closer to the ideal quality standard. ■
for the entire manufacturing process, including process design.

Finally, another, less imposing hurdle is the fact that the equip-
ment used in continuous manufacturing is both smaller and

24 www.patheon.com ● doingbusiness@patheon.com
Patheon
OneSourceTM

Single-Vendor
CDMOs Bring
Speed and
Cost Savings
to the Table

Anil Kane, Ph.D., MBA Paul Nelson, MBA


Global Head of Robert Fry, Ph.D. VP of Supply Doug Johnson
Technical & Scientific Chief Economist, Chain and R&D, President,
Affairs, Formulation, Robert Fry Amring OCAM
Patheon Economics, LLC Pharmaceuticals Solutions, Inc.

As drug developers face the ever-pressing need to get mol- Unfortunately, a multi-vendor approach often adds more
ecules to market as efficiently as possible, firms large and time and effort than many firms would like. Drug developers
small are increasingly turning to CDMOs for help. The need to negotiate numerous vendor contracts, complete
CDMO industry is evolving to meet this need, spurring some technology transfers, conduct revalidation, and complete
larger providers to offer forward-thinking services in the other time-consuming tasks.
form of end-to-end supply chain models for pharmaceutical
and biopharmaceutical clients. Compare this approach to a single-source vendor option,
which has the unique ability for both drug substance and
At a panel discussion held at CPhI North America 2017 in drug product teams to collaborate from development through
Philadelphia, four industry experts discussed how working commercialization, representing a major shift in the drug de-
with a single-source CDMO partner can accelerate time to velopment world. Aligned services in end-to-end supply
market, add cost savings, and improve a formulation’s chains include the development and optimization of drug
chances of achieving regulatory success. substances and products; the manufacture of supplies in
batch through commercial sizes; clinical and commercial
packaging; supply chain management; and more.
Speed and the single-source model
Because cross-team collaboration allows for real-time feedback
According to Anil Kane, Ph.D., MBA, Global Head of Technical about both the drug substance and drug product, one can move
and Scientifiec Affairs of Formulation at Patheon, speed is es- efficiently from an API to a drug product with fewer errors. Potential
sential in drug development. Patients need their medications, problem areas (like poor solubility, bioavailability, or process-abili-
and competitors are vying to get their products to market first. ty) can be worked out before the process is scaled up. If formula-
Working with even more compressed timelines are firms racing tion problems are caught in early development phases and work is
toward commercialization with life-saving therapies that have completed to understand the molecule and its characteristics, de-
fast-track status. Likewise, small biopharmas deal with incredible velopers will benefit from more robust process development that
time pressure as they function in a market-driven environment. avoids costly and time-consuming “re-dos” during scale-up.

www.patheon.com ● doingbusiness@patheon.com 25
Other time-savings that surface when all aspects of a proj- starting a new contract with another vendor. Rather, the sin-
ect lie with the same vendor are those associated with gle-source vendor was responsible for figuring out how to
external technology transfers, vendor qualification, negotia- remedy the problem with the API because it was their work
tions, and follow-up. All these activities are eliminated on to begin with.
the client end. According to one CDMO, its trademarked
single-vendor network “eliminates 8-12 weeks of develop- A single vendor with involvement in a project from soup to
ment time for small molecules and 14-20 weeks for large.”1 nuts has a unique opportunity to help their client by integrat-
ing information that is generated from all points along the
While the single-source CDMO option may well be a for- project continuum. “A single vendor can leverage expertise
ward-thinking approach in the world of pharmaceuticals and typically stashed in silos across different stages of the pro-
biopharmaceuticals, there is a strong precedent for it in oth- cess, from API development to formulation and the supply
er manufacturing sectors. For instance, Robert Fry, Ph.D., chain,” noted Kane.
Chief Economist at Robert Fry Economics, LLC, explained
that Toyota’s lean manufacturing model limited the number For example, in the Patheon OneSource™ model, a single
of suppliers it worked with so that the car maker could pro- program manager oversees all activities, and bridges vari-
duce new models much faster than its competitors. ous teams and sites performing the work. Having a program
manager so close to the client and the operations brings
efficiency and speed to the process.
Breaking down barriers: Communication in
Nelson agreed that this type of program management strat-
a single-source model egy can be key to a project’s success. Decisions on sourc-
A single vendor offers access to a network of experts across ing of API development and subsequent manufacturing are
several disciplines who can share knowledge about a proj- usually made before an application for regulatory approval
ect as a molecule moves from phase to phase, thus helping is filed. Many companies do not have the coordination need-
to ensure it stays on a path toward commercial success. ed to determine where final commercialization will be done
at the time they are getting ready to file. A program manager
Paul Nelson, MBA, Vice President of Supply Chain and R&D can facilitate good coordination and look at the value stream
at Amring Pharmaceuticals, offered an example of how hav- that cuts across all business units and functions.
ing such knowledge through a single vendor could have
benefited a recent R&D project he worked on. Several mol-
ecules in his pipeline had development work completed in Due diligence: Selecting the right partner
India while the manufacturing activities were conducted in
When helping pharma clients evaluate single-source ven-
France. The project came to a halt after the first pilot batch-
dors, Johnson said he looks for four must-have characteris-
es were made poorly. An investigation was launched to de-
tics: capability, flexibility, stability, and caring.
termine the source of the problem, probing the possibility of
a development issue, tech transfer problem, manufacturing ●● A variety of capabilities are essential for helping to im-
issue, or something else. The time-consuming process prove a process and elevate a compound from ear-
caused a significant delay. ly-stage work through commercialization. When trying
to make a good match between what a client is looking
If the development and initial manufacturing work had both
for in an outsourcing partner and the CDMO’s capabil-
been done by a single vendor, the problem may not have
ities, Nelson added that the process is as much an art
occurred. But if it had, the open channels of communica-
as it is a science. Several CDMOs may all have similar
tions that a single provider offers would have made identify-
equipment, but what really differentiates them are the
ing the root cause of the issue much easier.
dedication and knowledge of the individuals working
Doug Johnson, President of OCAM Solutions, Inc., adds on the projects. Is that know-how and capability em-
that communication in a single-vendor network helped fix a bedded in the organization? In some cases, clients
major problem his client had with an API. Had the client have been burned by the claim that an outsourcing
used multiple vendors, resolving the issue would have re- partner had competencies in areas it did not.
quired breaking a commitment with one manufacturer and
●● Without fail, unexpected twists and turns will surface
during drug development. A CDMO’s flexibility is criti-
cal, especially in early phases. Firms should have the
1 Patheon OneSource™, www.patheon.com/onesource/index.html, accessed
June 1, 2017.

26 www.patheon.com ● doingbusiness@patheon.com
dexterity to address unanticipated problems quickly When negotiating a contract with a single supplier, it is criti-
and adjust. cal to get the language right. From the stance of an econo-
mist, Fry said incentives must be balanced such that both
●● The vendor cannot simply provide a service; they must parties benefit from doing the right thing. It is crucial to con-
have a track record of providing services well. CDMOs sider all contingencies and to cover all the bases. For
tend to acquire other companies with expertise in a example, is the client protected if the price of raw material
specific area. A single-source vendor must also be re- sharply increased?
liable and stable — fiscally and in terms of longevity,
all while putting out high-quality work. Nelson noted that risk management is also important when
looking at an end-to-end supply chain. There is economic
●● Larger CDMOs may be working on hundreds of proj- risk, the change of a supply interruption, and even location
ects simultaneously, but a good partner also ensures risk, for instance, if the vendor is in a hurricane zone. All
every client receives the attention it deserves. types of risk must be examined to build the best risk-man-
agement strategy.
If a client pays more to have these four elements in a single
provider, Johnson said it is money well spent in exchange
for time saved and a better end-product. Summary
Like any new technology, some pharmaceutical companies
Streamlining from the start: are still hesitant to adopt the single-vendor sourcing model,
One vendor, one contract especially small, emerging firms. While some companies are
taking a watch-and-wait approach to see how the single-ven-
A key advantage of the single-vendor network is that clients dor concept pans out, those that are already embracing it are
will only have one contract. Having to negotiate contracts reaping the benefits of efficiency and expertise. ■
and legal agreements with multiple vendors requires a lot of
time and energy — even if just two suppliers are involved.
Nelson said he has seen contract negotiations between two
vendors working on the same project take as long as
18 months to finalize. This, of course, takes time away from
doing more valuable tasks, thus the single-source provider
is far more efficient in this respect.

www.patheon.com ● doingbusiness@patheon.com 27
Patheon
OneSource™
4 Perspectives
on Single-
Vendor Drug
Development
Models

Bringing the Single-Vendor Approach


to the Pharma Industry

Robert Fry, Ph.D.


Chief Economist,
Robert Fry Economics LLC

From the perspective of an economist, time is money. And, partic- single-supplier production process, which was an innovative part
ularly in the pharmaceutical industry, speed is of the essence. As of its lean production system. The single-source network was
drug developers face increasing pressure to get their products to mainly implemented for the purposes of better inventory man-
market as quickly as possible, the CDMO industry is evolving to agement, but it also enabled Toyota to create faster turnaround
meet this need by offering end-to-end services. In a recent inter- times than its competitors. Toyota could launch a new model on
view, Robert Fry, Ph.D., who, as a macroeconomist and Chief the market every four years while other companies were doing it
Economist at Robert Fry Economics LLC, offered his observations every 5 to 10. Toyota was faster because it was working with
on these important trends. fewer vendors; therefore, the car maker spent much less time
negotiating contracts and eliminated many logistical problems.

Q: What are the benefits to a Speed is also an important factor for the pharmaceutical indus-
pharmaceutical company (large or small) try. In the context of drug development, where competitors may
adopting a single-vendor approach? be coming out with similar products or patents are expiring, com-
panies want to get their products on the market as soon as
Fry: For the pharmaceutical industry, speed is an attractive possible. Speed is of the essence, which is perhaps the best ar-
benefit. To my knowledge, Toyota was the first company to use a gument for using a single supplier in the pharmaceutical industry.

28 www.patheon.com ● doingbusiness@patheon.com
Q: During the panel discussion at CPhI The longer the wait between when the money is expended and
when a return on it is realized, the more it costs. In the context
North America 2017, you described the
of drug development, more money is probably spent on good,
single-supplier approach as a middle solid research than on actual materials. In this scenario, what is
ground between two ends of a spectrum. really being paid for is human capital. In other words, the value
Why do you feel that’s the case? of the materials is secondary to the brain power that knows
what to do with them.
Fry: The single-supplier model is a middle ground between one
extreme, where there are many suppliers in a competitive indus-
try. At this end of the spectrum, it’s an options market, where Q: When a drug maker is evaluating these
products are bought from the cheapest vendor. In this scenario, types of contracts, what are the most
the item is a commodity, multiple suppliers are producing it, and
many consumers are buying it. The other extreme is an integrat- critical ingredients?
ed company that both produces and sells the product.
Fry: When building a contract, expertise is needed from many
In some industries, a single supplier is desirable whereas in different areas. It is important to involve an economist or busi-
others, an options model is sufficient. In the pharmaceutical ness person. The incentives must be structured such that both
industry, having a single supplier (i.e., the middle ground) may parties in a contract are acting in their combined best interest.
work better than having multiple suppliers (the options model) Good lawyers are required to cover contingencies. It is also
or doing everything in-house (i.e., the integrated approach). important to involve personnel from the technology side. In the
Drug development, which is research and labor intensive and context of drug development, it is not a case of simply minimiz-
involves products that are very specific to the buyer, lends itself ing the price of materials; the contract should be approached
to the single-supplier model. much more holistically.

From an economist’s point of view, there should always be a


Q: From the perspective of an economist, clause in case the cost of a material increases dramatically. For
what is the monetary value of using a example, if the price of a material doubles, the pharmaceutical
single supplier? client must agree to pay more for it. Otherwise, no one will sup-
ply it because it’s too risky. Including ways to cover contingen-
Fry: Time is money. Economists talk about a discount rate, or cies, such as price index clauses, are important to ensure that
the capital that is tied up in the rate of return paid on capital. risks are shared by both parties. ■

New Resource Saves Drug


Developers Time and Money

Anil Kane, Ph.D., MBA


Global Head of Technical and Scientific Affairs,
Formulation

In the pharmaceutical industry, speed is of the essence. As maceutical and biopharmaceutical clients by accelerating time
drug developers face increasing pressure to get formulations to to market, adding cost savings, and putting formulations on the
market as efficiently as possible, the CDMO industry is evolving path toward regulatory success. Here, Anil Kane, Ph.D., MBA,
to meet this need. The Patheon OneSource™ model exempli- Global Head of Technical and Scientific Affairs of Formulation
fies the benefits of an end-to-end supply chain model for phar- at Patheon, elaborates on his views on the subject.

www.patheon.com ● doingbusiness@patheon.com 29
By accelerating the development ability, and, thus, its probability of success. Patheon was able to
significantly improve the solubility of the compound by making
process, a single vendor can a solid dispersion, which resulted in better exposure.

deliver better healthcare to If the drug substance had been kept in-house, Patheon could
have applied its solubility-enhancement programs at an earlier
patients faster and bring stage, thus saving the client time and money.

business benefits to the sponsor.


Q: What concerns do prospective clients
have about the single-vendor approach?

Q: What are some top benefits of working Kane: The industry is still learning about the single-vendor
approach. However, many companies have experienced chal-
with a single-source CDMO? lenges with managing multiple vendors. A lot of time and
resources are required to oversee projects that are done at var-
Kane: We believe there are several key advantages when a
ious sites, which has spurred many large pharma companies to
CDMO aligns its drug substance and drug product units. Be-
reduce the number of vendors they work with.
cause a single-source network encompasses the entire process
from end-to-end, a single program manager oversees a client’s There are also benefits to having a single legal contract and
project from its early preclinical stages, through to commercial quality agreement. A lot of time is spent negotiating terms and
supply. A single-vendor CDMO ensures continuity, accelerates conditions with multiple vendors. Having to negotiate only one
scale-up, and helps set the stage for regulatory success. contract with a single vendor has benefits for companies that
do not have the time and resources to audit multiple vendors.
During development, strategies often change as the result of
clinical outcomes or regulatory requirements. With a sin-
gle-source network like Patheon OneSourceTM, the pathway is Q: Does the single-vendor approach have
nimbler than what a multi-source network offers, and can quick-
ly address issues that surface at any point from the preclinical
benefits regardless of a company’s size?
stage through clinical development and commercialization.
Kane: The pharmaceutical industry has traditionally worked
In addition, working with a single vendor throughout a product’s in silos with separate drug substance and drug product
lifecycle eliminates the need for expensive and time-consuming groups. Companies with a much smaller team may not have
technology transfers. It also ensures no knowledge is lost the necessary background to support the entire process from
during a transfer to a third party. drug substance to drug product, making the single-source
vendor quite advantageous.
Any delays in bringing a product to market mean lost opportuni-
ty from a business perspective and lost benefits to patients. By In addition, resources have been shrinking at medium and large
accelerating the development process, a single vendor can de- pharma companies, which are consolidating to focus on higher
liver better healthcare to patients faster and bring business priority products and portfolios. They are also reducing the num-
benefits to the sponsor. ber of staff members that manage outsourced products and ser-
vices. Thus, the single-vendor concept is becoming increasingly
beneficial to medium and large pharma companies, too. ■
Q: Do you have a case study that illustrates
the benefits realized by a small startup
company working with a single CDMO?

Kane: Patheon developed low-kilogram quantities of a cli-


ent’s drug substance for preclinical toxicity studies. The client
then took the compound to another vendor for early-stage for-
mulation. They tried several strategies that failed. The
compound was poorly soluble and did not attain the needed
exposure in animal models or Phase I clinical trials.

Two years later, the client asked us to revisit the compound.


They needed a strategy to improve its solubility and bioavail-

30 www.patheon.com ● doingbusiness@patheon.com
A Single-Vendor CDMO Eases
Communication Flow

Paul Nelson, MBA


Vice President of Supply Chain and R&D,
Amring Pharmaceuticals

A single vendor offers access to a network of experts across sev- Several risks must be managed to ensure that medicine gets to
eral disciplines who can share knowledge about a project as it patients consistently. I look at all the risks along a supply chain
moves from phase to phase, thus helping to navigate its path to- such as staff turnover at the vendor, financial issues, or location if
ward commercial success. In this article, Paul Nelson, MBA, Vice the firm is in a hurricane area, for example. If a single vendor can
President of Supply Chain and R&D at Amring Pharmaceuticals, show that this model works by reducing the risks associated with
discusses his experience with single-source CDMOs. clinical and commercial supply, I think the market will embrace it.

Q: What does it take for a single-source Q: What is the most important thing to know
CDMO to be successful and to overcome about a vendor before signing a contract?
the silo mentality that is often found in
Nelson: The vendor must have the expertise to deliver on its
pharma companies? promises. Contracts are developed on the basis of assumptions
about how well the CDMO will perform. The decision to sign on
Nelson: I had an experience where the silos did, in fact, the line involves more than just a good PowerPoint presentation.
show up in a single-vendor model. In that case, having one There must also be tangible experience that gives the sponsor
CDMO didn’t overcome the silo mentality, and the partnership confidence that the CDMO will meet the outlined expectations.
didn’t work very well.
For example, it is important to know who works for the vendor
For the single-vendor model to work, the CDMO must have a (that is, who are the subject-matter experts), and there must be
single project management strategy throughout the relationship some level of confidence that they are going to stay. It would not
from beginning to end. That way, the client doesn’t have to work be ideal to sign a contract with a vendor on the basis of exper-
with one part of the CDMO for raw materials and API develop- tise that is in place at the time, only to have those key personnel
ment, another group for conversion to finished product, and yet leave the company soon thereafter. In that case, the client is left
another unit for packaging. It’s a more streamlined and suc- hoping that the vendor’s know-how has not left with the people.
cessful approach to drug development.

Q: What risk management issues must be


For the single-vendor model
resolved for a pharma company to fully to work, the CDMO must
embrace the single-vendor model?
have a single project
Nelson: It depends. Risk management is improved if the
CDMO has been in business for a long time, is financially sta-
management strategy
ble, has a well-established management team, and possesses throughout the relationship
the know-how to do the work from the beginning to the end of
product development and supply. from beginning to end.
www.patheon.com ● doingbusiness@patheon.com 31
Q: Are there other benefits to working with Q: If a larger pharma company has a lot of
a single vendor from preclinical product in-house capabilities, including a good
development to supply? preclinical/early development team, is there a
benefit to outsourcing early in the process?
Nelson: In addition to silos, there are barriers to
communication between organizations. I’m working on
several products that are being developed in one country,
Nelson: In the case of larger companies, the most benefit
from having an outsourcing partner comes in Phase III and
and scaled up and packaged in others. Getting the different
languages, cultures, and priorities of multiple companies in beyond. However, mainly from a knowledge-transfer stand-
sync and maintaining it from preclinical development through point, there could be some benefit to having the same
finished product is very time consuming. A lot of resources CDMO that is going to provide the final medicine be involved
are required to manage cooperation among different early on. For example, it could be useful for the CDMO to
organizations. In this respect, working with a single vendor have in-house, first-hand knowledge of how the compound
would have numerous advantages. behaves in the laboratory before the transition to scale up. ■

Smaller Biotech Companies


and the Single-Source CDMO

Doug Johnson, Ph.D.


Founder,
OCAM Solutions

A single-source CDMO that manages the end-to-end relation- of two strong, capable companies brings advantages in time-
ship with pharmaceutical or biopharmaceutical clients can bring line synergies, information flow, and general project integration
many benefits and efficiencies to the development process. without having to sacrifice competency.
However, smaller companies, in particular, may still be reluctant
to work with a much larger, single-source vendor. Expanding
upon the discussion, Doug Johnson, Ph.D., Founder of OCAM Q: What might be a pharmaceutical or
Solutions, points out these important issues. biotech company’s biggest concerns
with using a single-source vendor?
Q: During the panel discussion at CPhI North
America, you stated that the value of the Johnson: I work mostly with small companies, which are
sometimes concerned that they won’t get the equivalent amount of
single-vendor model is increasing. How so?
attention from the contractor as a large pharma company. This is
Johnson: How vendors have developed competencies has less of an issue when a small pharma/biotech company is working
changed over time. More recently, single-source contractors with a small contractor, because in that scenario, the client gets to
have been expanding by acquiring very competent companies know the executives at the contract organization. When larger
rather than by trying to cultivate a particular competency in- CDMOs are created through acquisitions, it is not always possible
house (i.e., without the starting point of capability). The merging for clients to get that personal touch from the top.

32 www.patheon.com ● doingbusiness@patheon.com
I work mostly with small In another case, our client did not initially choose to work with a
large contractor. The vendor they contracted to manufacture their
companies, which are drug substance was subsequently acquired by a larger company.
The transition was managed well and didn’t create any glitches for
sometimes concerned that the client. They were so pleased with the work done by the drug
substance group that they opted to stay with the company for their
they won’t get the equivalent drug product work. Even though the client didn’t initially plan to use
a single vendor, it worked out well.
amount of attention from
the contractor as a large Q: Are there advantages in terms of
when clients begin working with a
pharma company. single-source CDMO (e.g., in Phase III
Also, large companies sometimes have the reputation of being versus early formulation)?
more policy-driven than science-driven, or of being more bureau-
cratic and less flexible. Small companies have an affinity toward Johnson: There has been a shift on the part of larger contrac-
working with contractors that they perceive to have modus operan- tors to being more interested in working with clients from the begin-
di similar to their own (e.g., being highly flexible and entrepreneur- ning. I have two clients for whom this has been particularly beneficial.
ial). This concern is allayed if a company has a good experience In one case, the project progressed as far as it could and was put on
with a large, soup-to-nuts contractor. If the contractor has to make hold. However, because the same company worked on both sec-
changes on the fly and proves to be responsive and flexible, then tions of their project, it means there will only be one source of data
the small company will be more willing to sign a contract with them when they have to perform due diligence, which will be cleaner and
for their next project. easier. The other case involved steps in which there were delays.
Managing delays in the process is much easier when both the drug
substance and the drug product are worked on within the same
Q: Do you have any specific examples of company because a delay in one company doesn’t have to be ex-
how a single-source CDMO may benefit a plained to another. If one company is involved from start to finish,
small start-up company? the client’s project has the advantage of being less fragmented. ■

Johnson: In one case, a client selected a drug substance


manufacturer because of its unique fit between what the vendor
had to offer and what the client needed. This was for a Phase I/II
clinical supply run. The manufacturer was also able to produce the
For more perspectives on
active pharmaceutical ingredient (API) in capsules, which was ex- single-vendor vs. multi-vendor
actly what the client needed. A lot of hassle that would have been
associated with method transfer was avoided, because the meth-
manufacturing approaches,
ods were very similar. In addition, the timing was more coordinated visit patheon.com/onesource.
between production of the API and production of the drug product.

www.patheon.com ● doingbusiness@patheon.com 33
Clinical
Services

Cold-Chain Fully
Automated
Assembly and
Labeling of
Pre-Filled Syringes
for Clinical Trials

Cold is undeniably the new normal for the biopharmaceutical automated assembly and labeling of pre-filled syringes under
industry. By 2020, greater than half of bestselling drugs will be cold (2° C to 25° C) or ambient conditions available. Introduced
cold-chain products, most of which are injectable.1 in 2017, this service is exclusively available at the Basel, Swit-
zerland facility, where a dedicated cold facility maintains cold
With sponsors devoting much of their resources to developing
room conditions from truck to dock, through assembly, storage,
these large molecule products, the demand for pre-filled syringes
and distribution.
is escalating rapidly. Many large biopharma companies have their
own dedicated automated facilities for assembling and labeling
pre-filled syringes for commercial products. However, some lack Accessing automated solutions
sufficient capacity to meet both commercial requirements and the
growing demands of global clinical trials. Efforts to address both Placing tablets into blister packaging is simple compared with
needs can lead to time-consuming bottlenecks as commercial assembling and labeling pre-filled syringes.
and clinical operations compete for finite internal resources.
As recently as five years ago, help from a clinical supply chain
In an effort to forestall bottlenecks and conflicts, sponsors are partner in assembling and labeling pre-filled syringes for a clin-
turning to outsourcing partners for help. The process of assem- ical trial meant a painstakingly time-consuming manual pro-
bling and labeling prefilled syringes is considerably more chal- cess. This limited the use of pre-filled syringes to relatively
lenging than that of inserting tablets into blister packs, due to small trials or production runs. Thankfully, times have changed,
the special handling requirements of biologics. and so have the options.
To protect the shelf life of costly biologics, these products re-
To illustrate the difference between then and now, imagine a
quire refrigeration, with time out of the cold chain — also known
production suite containing 10 trained operators who spend a
as time out of environment or TOE — sharply limited. In fact, as
day manually assembling and labeling pre-filled syringes. At
many as 40% of biological products in development today are
the end of the shift day, the yield would be about 400 syringes.
permitted zero time out of environment, meaning that the entire
process of assembly, labeling, and packaging of pre-filled sy- Today, using precision automated equipment in that same pro-
ringes must take place in cold rooms. The proportion of these duction suite, the yield at the end of the shift day is about
biologics requiring 24/7 refrigeration is projected to rise.

To address this growing need, Fisher Clinical Services has be- 1 “2015 Biopharma cold-chain forecast.” Pharmaceuticalcommerce.com.
Pharmaceutical Commerce. 19 December 2016. http:// pharmaceuticalcom-
come the first supply chain partner in the industry to make fully merce.com/special-report/the-2015- biopharma-cold-chain-landscape/

34 www.patheon.com ● doingbusiness@patheon.com
13,000 syringes. This tremendous productivity increase makes Innovative automated solutions
it practical and cost-effective to use pre-filled syringes instead
positioned globally
of higher-priced vials and needles for studies involving thou-
sands of subjects. As the use of pre-filled syringes continues to grow, flexibility
and careful planning in clinical settings will benefit sponsors
during development and beyond by establishing a strong foun-
Quality assurance: Consistency and control dation for a commercial plan. Meanwhile, suppliers must adapt
to the changing needs of sponsors by continuously innovating
Precision automation also offers quality assurance that is supe-
new solutions.
rior to that of manual assembly when conducting large runs,
making the use of pre-filled syringes as risk-free an experience So rapidly has demand grown that Fisher Clinical Services now
as possible for healthcare professionals and patients. offers precision pre-filled syringe assembly and labeling at
three of its facilities — in Allentown, PA, U.S.; in Basel, Switzer-
Variations in the manual assembly of pre-filled syringes are in- land; and in Horsham, U.K. All three facilities employ a consis-
evitable and can affect performance. For example, an under- or tent global process. This global expansion of services include
over-turned plunger rod can lead to leakage that compromises eliminating glass-to-glass and glass-to-steel contact in equip-
syringe integrity, ruining the unit, and wasting costly IMP. ment. Glass-to-glass contact can cause syringes to crack,
causing safety issues, compromising product sterility, and re-
Similarly, an imprecisely applied needle safety device (NSD) sulting in waste. Glass-to-steel contact can scratch syringes, a
could result in a needle stick injury, an unnerving experience cosmetic effect that can nonetheless cause units to be rejected.
even for a healthcare professional who knows the syringe was
not previously used. The latest innovation includes the labeling and secondary packag-
ing of pre-filled syringes by fully automated, quality-driven, pro-
Automated assembly and labeling de-risk the process through cesses and technology under both cold and ambient conditions.
consistency and control. Automated systems vary, but the best
ones are highly precise and efficient, assembling and labeling Introduced in 2017, the fully automated, cold chain services are
pre-filled syringes in one fully automatic process. The systems currently available at the Basel facility, while both the Allentown
and Basel facilities support fully-automated operations. Semi-au-
employ a combination of customized equipment, sensors,
tomated services continue to be performed at Horsham.
torque control, and camera systems that permit operators to
maintain full control over the entire process from start to finish. Like all Fisher Clinical Services’ options for assembling and la-
beling pre-filled syringes, the patent-pending, cold-chain
For instance, a fully-automated compliance check uses a cam-
system in Basel is designed to meet the specific, unique re-
era system to detect the position of the syringe at all times. quirements of clinical trials. Here are the key benefits:
Sensors monitor every step of the assembly and labeling pro-
cess, reading exactly how much force is being applied to a ●● Reduces IMP wastage
backstop or plunger, for instance, and thus eliminating the risk
of under- or over-turning. Using camera checks, a labeling plat- ●● Mitigates TOE via cold room assembly
form ensures that the correct patient label is affixed to the cor- ●● Maximizes flexibility to meet the needs of every trial and
responding syringe. every syringe
Automated assembly and labeling of pre-filled syringes are in- ●● Ensures accuracy and legibility of fixed and variable text
creasingly preferred by most sponsors, but manual assembly
may be the most flexible and cost-effective option for small tri- ●● Safeguards patients and medical staff
als or production runs. New options are now available that
●● Mitigates risk through global processes and quality
couple some elements of automation with manual assembly to
deliver greater precision to smaller runs. ●● Delivers efficiencies and eliminates costly bottlenecks. ■

Sponsors can benefit from these options by building flexibility


into their planning and by exploring the assembly and labeling
services that are most suited to their needs. It’s important to
engage a supplier that is capable of meeting both automated
and manual needs.

www.patheon.com ● doingbusiness@patheon.com 35
Commercial
Manufacturing
and Technology
Transfer

Considerations
in Selecting an
Outsourced
Solution

Gavin Murdoch
Senior Director, Strategic Partnerships
Business Development

The traditional business model for in-house pharmaceutical chemical or biologic process. However, a small company may
manufacturing is nearly a thing of the past. More companies are choose to outsource this stage virtually. Either way, most of
turning to outsourcing to achieve flexibility and efficiency in a the focus at this point is getting the molecule to the clinic with
highly competitive market. According to a recent report by busi- little consideration for future scalability or its commercial sup-
ness intelligence provider Visiongain, the global contract bio- ply. It is in Phase II clinical development when manufacturers
pharmaceutical manufacturing market is predicted to reach often consider more advanced formulation development.
$79.24 billion in 2019. This is a substantial increase from
$54.54 billion in 2013.1 Nevertheless, this does not mean an out- For those companies that have been working virtually to de-
sourcing strategy is necessary for every project. A thorough velop a product and formulation, Phase II can be the ceiling
evaluation of a manufacturer’s capabilities and true cost struc- for scalability with existing partners. Typically, smaller-scale
ture must be completed early to determine if its goals can be virtual providers are not capable of scaling up or may not
achieved in-house. More importantly, this decision must be made have commercial good manufacturing practice (GMP) com-
early to allow enough time to initiate an outsourcing partnership. pliance. It is at this stage when small companies must start
to consider outsourcing for registration and commercial
supply for their project. For larger pharma companies, the
Expertise and capacity:
traditional perception is that, because they have the neces-
Do you have what it takes? sary capabilities in-house, it is best to keep manufacturing
The drug development process begins with discovery, where there. Yet, given the complexity of new products, past man-
most companies rely on in-house scientists to create the ufacturing capabilities and technical know-how may not be
adequate, and outside options should be considered. CD-
MOs may have a strategic advantage based on their expo-
1 Visiongain, Pharmaceutical Contract Manufacturing World Market to Reach sure to a variety of projects with multiple types of processes,
$79.24BN in 2019 — https://www.visiongain.com/Press_Release/761/%E2% and that type of experience can translate to a higher level of
80%9CPharmaceutical-Contract-Manufacturing-World-Market- To-Reach-
79-24bn-In-2019%E2%80%9D-New-Visiongain-Study-Predicts knowledge in both operations and development.

36 www.patheon.com ● doingbusiness@patheon.com
Capacity is also a factor at this stage, as a company must ing participated in the initial selection of the partner. They
ensure it has the space to manufacture its product. Demand often bring new questions and new needs that can compli-
forecasting for the future commercial product and, in partic- cate the deal and delay the project. A company can prevent
ular, launch forecast accuracy are often significantly wrong. this kind of delay by ensuring all appropriate parties are in-
In fact, over 60% of drug forecasts over- or underestimate volved from the beginning rather than handing off to
peak revenues by more than 40%. 2 These inaccurate fore- Procurement or Supply Chain to manage exclusively. Final-
casts have damaging and costly impacts. When demand is ly, the deal terms and conditions and scope for the project
overestimated, a decision to manufacture in-house results will be formalized in a master service agreement (MSA).
in expensive, unused facility capacity for both drug sub- Even in a scenario where there is an attempt to expedite this
stance and drug product. When demand is underestimated, process, it is best to anticipate a three- to six-month waiting
the manufacturer faces a race against the clock to quickly period to complete this part of the process. New stakehold-
produce lifesaving medications that must still be of the high- ers from legal, finance and supply chain are often involved
est quality. This takes place while facing lost sales. here for the first time, and the process of each trying to pro-
tect the interests of their company can affect the original
In addition to standard capacity needs, it is important to need and timeline. It is common for companies to not fully
consider any specific or unique technologies needed for the understand their own negotiation and approval processes,
product. New and emerging technologies, including sterile leading to rework and delays, depending on the size of the
single-use processes, biologics perfusion, HME or continu- companies and number of people involved.
ous manufacturing, create exciting possibilities for improved
development and commercial efficiency and flexibility. As Making a decision between keeping a project in-house ver-
these technologies are not as widespread, they can be diffi- sus outsourcing is not an easy one, especially with so much
cult to apply internally for just a single project or product. at stake. If a company has the expertise, technology, and
When it comes to expertise and capacity, a manufacturer experience necessary to work with today’s most innovative
must consider if it wants to invest in the equipment and ex- drugs, it may have a case to stay in-house. However, if it
pertise needed for these technological approaches; or if it does not have those capabilities, evaluation of an outsourc-
wants to defer to a CDMO with greater experience and a ing solution will provide the optimum long-term solution. By
variety of projects to stabilize the expertise. working back from your desired launch date and including
the steps above in your development and manufacturing
timeline, you will be able to better manage the process in
Timeline considerations order to meet your goals. ■
after partner selection
Companies that ultimately decide to outsource often under-
estimate the time required once the decision has been
made. The supplier selection process, including requests
for proposals (RFPs), evaluation of capable suppliers, and
final selection, takes considerable time and is often man-
aged by groups far removed from the technology experts.
This step can take up to six months to complete, time which
becomes critical at start up. A quality audit should be part of
a CDMO evaluation, which, depending on availability of in-
ternal and/or external quality experts, could require a wait-
ing period of up to three months.

After selection, a kickoff and onboarding process with the


selected partner is necessary, including the commitment of
stakeholders. It is not uncommon at this point for research
and operations personnel to become engaged after not hav-

2 Patheon, Implications of Inaccurate Forecasting on Biologics Drug


Substance Manufacturing – White Paper — https://www.patheon.com/en-us/
Ideas-in-Action/Material-Details/59/Implications-of-Inaccurate-
Forecasting-on-Biologics-Drug-Substance-Manufacturing---White-Paper

www.patheon.com ● doingbusiness@patheon.com 37
Commercial
Drug
Manufacturing
Product
and Technology
Services
Transfer

Technology
Transfers:
Reaping
Rewards,
Reducing
Risks

Thomas Dax Mirko Gabriele Paul Jorjorian


Director, External Global DPS Technology Transfer, Director,
Manufacturing, API Senior Manager Global Technology Transfer

There are a number of strategic advantages companies can However, Patheon has a great deal of experience in technology
achieve in pharmaceutical production by transferring production transfers and has developed processes and techniques for
between sites. They can safeguard supply by producing at more reaping its rewards while reducing its risks.
than one site, and improve distribution by moving production clos-
er to critical markets. They can also reduce program costs and
risks by moving production to sites that are better qualified, can When it works: Three successful transfers
produce more economically, or are better positioned to meet the
needs of regulators. 1. Product X
For several reasons, transferring production — and the technol- The product was a registered intermediate of a novel first-in-class
ogies that undergird it — can be risky. The same product can API Patheon was producing for a client in a 10-step process. One
behave differently in different equipment, resulting in low yields high-temperature step was especially complex, requiring good
or even batch rejections; staff at a receiving facility may not have control to balance quality and yield. We were spending a dispropor-
the proper technical skills to execute a specific process; or there tionate amount of time and effort getting this step right, and we
may simply not be enough trained staff on site to assure that the knew that other manufacturers — with more experience in this spe-
job will be done correctly. Also, even as companies may not take cific technology — could likely do it better. Accordingly, we set out
the time (or expend the effort) to create and provide good docu- to find a manufacturer who could take over this production step for
mentation of their product or process, their partners may not in- us, and we found one in China that could supply our plant in Austria.
vest the necessary resources to ensure efficiency and quality.
To get the process up and running on the supplier site in China,
If any of the above occurs, time and money will be lost in reme- we sent two of our people to the Chinese plant to work with
diation and patients may not receive their medication. them for a week; from the first batch, the product was in spec.

The net of these risks is that companies do not transfer produc- Including four weeks to evaluate suppliers, four weeks of tech
tion as often as they probably should. transfer activities, and five-to-eight weeks for raw materials and

38 www.patheon.com ● doingbusiness@patheon.com
lead times, the total time from project start to the first batch IV. Manufacturability, which means looking at product issues
produced in China was about four months. and process robustness
V. Market, assessing the impact on volumes of competing
products, as well as levels of market acceptance, among
2. Product Y
other factors
This product was a recombinant-fusion protein entering Phase III VI. Measurement, which models the boundary limits of the
clinical trials. Patheon’s client had a partially-developed perfu- preceding five Ms
sion process and wanted production to commence simultane-
VII. Mitigation, in which we develop a thorough plan to man-
ously in Europe and Asia-Pacific. Their timeline was tight.
age the identified risks
We transferred the process from our client to our plant in A comprehensive risk assessment will yield a predictable set of
Groningen, the Netherlands, and our plant in Brisbane, activities on which a plan can be constructed. Such a plan al-
Australia, scaling up production in both at the same time and lows all stakeholders to begin on the same page, ensuring the
writing half the batch records in each location. alignment of both the sending and receiving teams before the
actual transfer. Furthermore, these steps generate thorough
The total time from receiving the process information and or-
documentation of both process and product, making knowl-
dering the raw materials to full production at both sites was four
edge transfer easier and less vulnerable to informational gaps.
months — about 30% to 40% faster than the industry average.
Once identified, those can be filled, and steps that need to be
improved can be identified (as was the case with Product Z’s
3. Product Z lyophilization cycle) and resolved before the transfer.

Product Z was a small-molecule, lyophilized product. Our client


wanted to transfer commercial production to Patheon. Unfortu- 2. Fixing problems prior to transfer
nately, the technical data we received about the product and
As noted, our assessment of product Z revealed a weakness in
the lyophilization cycle (the freeze-drying process), was not
the lyophilization (lyo) cycle, notably cake appearance issues.
sufficiently robust to guarantee a consistently high-quality prod-
Accordingly, and together with the client, we performed a sys-
uct. In particular, we could see from the historical data that the
tematic characterization of the product and its formulation to un-
lyophilizing cycle was not challenged at the edge of the critical
derstand better its behavior during lyophilization. We mimicked
process parameters (CPP), and there were several cake ap-
the lyo process in the lab, identifying the critical process param-
pearance issues even under optimum conditions.
eters that were likely to vary in production, such as equipment
Instead of replicating the process as it was, we worked with the performance, temperature, humidity, and time and velocity rang-
client to improve it, thereby getting the product right the first time, es. Then, in the lab, we measured the impact of those factors on
and improving both yield and quality over the prior process. yield, quality, cosmetic appearance, and varied them to see what
changes produced the highest quality across all measures. In
this way, we developed a more complete understanding of the
Why it works: A framework for factors that affected production and defined the envelope in
successful tech transfers which quality and yield would be optimal. During this process, we
also were able to understand better the physical characteristics
Through the examples of products X, Y, and Z, we can see four of the API, ensuring more reliable behavior in production, and
important elements of our approach in operation. limiting the impact of these variables on the drug product.

Fixing the lyo problem before the transfer enabled us to im-


1. Assessing risks and developing prove the process, strengthen the product’s regulatory submis-
mitigation plans prior to transfer sion with the additional information we obtained, and created a
strong relationship with our client that served us both well in this
To do that, we deploy a seven-step process we call the 7Ms. They are: transfer and in subsequent ones.

I. Machines, meaning their capacity, and the forecast analysis


II. Materials, taking into account such supply issues as lead time, 3. Standardizing processes and equipment
availability, quality, and whatever supplier issues we foresee
For the recombinant fusion protein, Product Y, Patheon’s plants
III. Manpower, including staffing requirements as they are af- in Groningen and Brisbane essentially had the same upstream
fected by demand fluctuations and downstream equipment (such as chromatography columns

www.patheon.com ● doingbusiness@patheon.com 39
and skids), so we could be confident that what worked in one Why companies are leery of
would work in the other. And not only was the equipment the
technology transfers
same, but the two plants also have very similar operating pro-
cedures. For example, each plant operates the 500-liter perfu- As noted, technology transfers don’t happen as often as they
sion bioreactor (a finicky process) in the same way and have might, or as often as perhaps they should. Companies have
almost identical standard operating procedures for common legitimate concerns about supply interruptions and the costs
downstream processes. Consequently, batch records written they may incur as a result.
for one plant could be transferred easily to the other.
Then there is the expense. To plan one thoroughly and execute
We also made sure that the materials and supplies were al- it carefully requires a significant investment. To minimize the
ready in both facilities’ systems, so neither they nor their suppli- cost, companies are sometimes tempted to shortcut the plan-
ers needed to be requalified, saving time and ensuring consis-
ning and rely on what’s worked before for similar products. This
tency and quality.
is a false economy. Looking at our history of transfers, we have
found that the chance of getting them right by mimicking the
4. Paying as much attention transfer of a similar product are no better than 50% — a coin
toss. Conversely, beginning fresh, with a blank sheet, and ana-
to people as to process
lyzing the process as if you have never done it before, the
Typically, the staff at the sending plant have expertise in a partic- chance of right-the-first-time success is better than 90%.
ular product and process. That, of course, should be captured in
Yes, technology transfers require investment. However, the
the documentation. However, even if it is, a specific answer to a
cost is less than that of fixing one that went wrong.
niggling problem may not be easy to find, and even with the best
documentation, a nuance may go missing. The staff at the re- In addition, a technology transfer provides the opportunity to
ceiving plant may have deeper expertise in the type of product,
make product and process improvements that might otherwise
or process, that can be applied to improve the existing process
be missed. In practice, production processes often run for years
and streamline the transfer. However, sender and receiver can
without being improved meaningfully. Companies always have
only help each other, and optimize the transfer, if their working
other priorities, including new projects, which steal attention from
relationship is close, collegial, and congenial. The single most
ongoing products. But in making a transfer, a company is forced
important ingredient that makes that relationship work is trust.
to make changes; there will always be differences at the receiv-
And trust emerges most reliably from successful collaboration.
ing site that need to be accommodated. Those changes — prop-
For the Fc-fusion protein, Product Y, the teams at each of the two erly planned for and executed — can realize enduring strategic
plants communicated in real time about progress, challenges, advantages by lowering production or distribution costs, or by
solutions, and so on. Joint project team meetings were held regu- improving a company’s competitive position in a given market.
larly, and communication between the plant managers was
excellent. Given that all the communication was done remotely, it
certainly helped that many of the people in the Groningen and
A challenge worth accepting
Brisbane plants knew and had worked with each other previously.
Transferring the production of sophisticated products from one
If teams have not worked together before, partners need to take site to another, or to several others, will always be challenging,
the time and trouble to allow trust to develop naturally through and will always involve risk. But not only are there more opportu-
proximity. For example, for the API production step on nities today for companies to improve how they have their
Product X that we transferred to China, we assumed the not in- products made, there are also more options for securing supply,
considerable cost of sending knowledgeable people thousands producing closer to critical markets, being more agile in respond-
of miles to the site. That was money well spent. It served to ac- ing to changes in demand, and securing sophisticated help with
celerate the knowledge transfer and when problems did surface technically challenging production steps. Specialist expertise,
(as they inevitably do), the receiving plant was not left to its own solid processes, and standardized operating procedures and
devices to try to solve or fix them without adequate support. equipment all work together to minimize risk and costs.

In the case of Product Z, we assembled a joint team with the cli- A consistent risk-based approach, continually updated with new
ent to work on improving the process, giving both parties time to data from quality by design and design of experiments initiatives,
get to know each other before the transfer. This paid off down the will improve both Patheon’s and the industry’s ability to efficient-
road as the team was able to work together productively through- ly, effectively, and seamlessly transfer production going forward.
out the lifetime of the product, supporting each other in matters The rewards can be substantial, and transferring technology
concerning strategy, supply, distribution, and regulatory affairs. should be an option every company places top of mind. ■

40 www.patheon.com ● doingbusiness@patheon.com
Client
Success
Stories

How Pacira
Pharma is
Working to
Help Curb the
Opioid
Epidemic

Poorly managed postoperative pain can have a significant impact this epidemic, the FDA assembled a task force in 2013, which,
on a patient’s recovery. For example, it can put them at risk for among other efforts, encouraged the development of abuse-deter-
medical complications, including deep vein thrombosis, pulmonary rent formulations of opioids. However, the most effective way to
embolus, and pneumonia.1 In addition, today’s methods for drug stop opioid dependence is to eliminate the use of these highly ad-
delivery often introduce a drug into the body in large quantities dictive drugs after surgery altogether. This was the goal of Pacira
where it must then systematically travel until it reaches the intended Pharmaceuticals, a specialty pharmaceutical company focused on
area. This wide-scale distribution increases the potential for harm- the development of non-opioid products for postsurgical pain con-
ful side effects and requires a higher cost for care due to the amount trol, when it developed its injectable suspension, EXPAREL®.
of drug needed.

Another concern about the impact of poorly managed postopera- Changing the standard of care
tive pain is related to the use of opioids for pain relief and a rise in
EXPAREL is a local analgesic utilizing bupivacaine in combination
the misuse and abuse of these drugs by patients. In a recent study
with the product delivery platform DepoFoam®. DepoFoam is made
from Stanford University, researchers found that patients undergo-
of microscopic polyhedral particles composed of numerous non-
ing 11 of the most common types of surgery were at increased risk
concentric internal chambers that encapsulate a drug. These dis-
for chronic opioid use.2 The increased use of opioids after surgery
crete chambers are separated by lipid membranes and filled with an
has caused a cascade into the abuse of prescription painkillers,
aqueous solution of a drug. Once the drug is inside the DepoFoam
and opioid dependence rose more than 3,000% from 2007 to
technology and injected into the patient, their body begins to break
2014.3 According to AddictionCenter.com, there are an estimated
down the lipid membranes. This is a naturally occurring process
4.7 million people in the U.S. dependent on painkillers.4 To address
that releases the drug without altering its molecular structure.

1. Agency for Healthcare Research and Quality, Improving the Quality of Care DepoFoam permits both systemic and local delivery, which means
Through Pain Assessment and Management — http://archive.ahrq.gov/
professionals/clinicians-providers/resources/nursing/resources/nurseshdbk/ the platform can release drugs into the bloodstream via the intersti-
WellsN_SMTEP.pdf tial space or into a body compartment, such as a joint. This target-
2. National Public Radio, Many More People Seek Medical Help For Opioid ed delivery system allows the injection of drugs into the areas of the
Abuse — http://www.npr.org/sections/health-shots/2016/08/01/487940139/
many-more-people-are-seeking-medical-help-for-opioid-abuse body where pain receptors exist, focusing the administration of
EXPAREL to only the areas where it is needed.
3. Surgical Products, Stanford Study Confirms Post-Surgical Patients Are
Especially Susceptible
The delivery of the non-opioid drug, EXPAREL, through the
4.To Opioid Abuse — http://www.surgicalproductsmag.com/news/2016/07/ DepoFoam technology provides physicians and patients with the
stanford-study-confirms-post-surgical-patients-are-especially-suscepti-
ble-opioid-abuse Addiction Center, Painkiller Addiction Statistics — https:// tools necessary to overcome the challenges associated with man-
www.addictioncenter.com/painkillers/

www.patheon.com ● doingbusiness@patheon.com 41
aging postoperative pain. “EXPAREL limits the amount of drug turing process into one of their suites, and then, within that work,
needed after surgery, and, as a result, the potential for adverse ef- expanding the process out and training their personnel on how to
fects is reduced dramatically,” says Ray Kaczmarek, Vice President run it. We will monitor the process as a partner as they manufacture
of Commercial Manufacturing and Supply Operations at Pacira. the drug product.” This is accomplished through the use of
“Most importantly, it empowers physicians to change the standard Patheon’s condominium manufacturing model. The “condo” model
of care for postsurgical pain and ultimately gets the patient back up is one of six adaptable manufacturing arrangements the CDMO
on their feet without the severe adverse events or the side effects of offers its clients. Through multiple options, Patheon provides spon-
an opioid.” After it is infiltrated into the surgical wound prior to clo- sors with a variety of flexible and scalable manufacturing approach-
sure, EXPAREL can deliver 72 hours of postsurgical anesthesia, es to create the optimal solution to fit a client’s needs.
Kaczmarek says. This significantly reduces pain during the first 24
to 48 hours after surgery, when it is experienced the most.
A stronger partnership through
Addressing the challenges of collaboration and communication
EXPAREL manufacturing Patheon’s condo model is designed for companies either expand-
ing their current manufacturing capacity or introducing new prod-
Beyond the composition of lipid components and the need for uct with unique characteristics that cannot be manufactured on a
aqueous excipients, the effectiveness of DepoFoam’s delivery plat- conventional manufacturing line, such as EXPAREL. In addition to
form is dependent on the manufacturing process used to develop
providing manufacturing design services, Patheon works with
it. Kaczmarek says one challenge is the critical need to properly
equipment suppliers, validates the processes, builds the line, and
handle EXPAREL in its sterile state. “Once we start the process of
manages operations on behalf of the client. Overhead is shared,
creating the liposome, we have to maintain sterility assurance all
and the line can operate as needed to meet demand.
the way through to the patient,” he explains. “That means the bulk-
ing process, the filling process, and everything done until the point Kaczmarek says Pacira personnel support the manufacturing and
where the physician begins to utilize the drug.” Other important quality assurance processes in Patheon’s U.K. facility. “This gives us
factors for the successful manufacturing of EXPAREL include tem- the ability to utilize the facility, the staffing, and the support structure
perature, agitation, and the design of the sterility assurance pack- Patheon brings to the table,” he says. “They also have multiple areas
age around the drug. outside of the U.K. facility where we can continue to grow strategical-
What also makes the manufacturing process for EXPAREL chal- ly.” Kaczmarek adds that Pacira wanted a manufacturing agreement
lenging is the need for a manufacturer to understand the complexity and strategic partner, not just a one-time partner for one product.
of the drug’s critical process parameters and how they interrelate to
The condo model design coupled with the manufacturing agree-
each one of its attributes. If any one of the parameters is changed,
ment gave Pacira a high level of confidence that they not only
the drug product will either fall apart or will not work in an appropri-
picked the right partner, but they would also be able to control the
ate way. Additionally, the limitations of today’s downstream technol-
intellectual property of their drug product design and formulation.
ogies create a manufacturing bottleneck that makes the scale-up of
“Other potential manufacturing groups we were looking at did not
EXPAREL difficult. “The sooner the industry starts improving down-
have the same type of model, nor did they have the same ability to
stream technologies, the sooner we will be able to maximize the
provide that type of an operational environment where we could
technologies, such as TFF [tangential flow filtration], to improve our
manufacturing scale,” explains Kaczmarek. “Until then, we’re creat- transfer a very complex process to them with the understanding of
ing newer platforms and utilizing different technologies to allow the how they would actually be able to manufacture moving forward,”
maximization of scaling by limiting exposure in the downstream pro- says Kaczmarek.
cess and implementing continuous batch processing.”
The next step toward creating an even stronger relationship,
It was these challenges, as well as the need for global exposure Pacira and Patheon are establishing a governance relationship,
and regulatory compliance, that caused Pacira to seek a partner where both companies speak on a regular basis and provide up-
who could provide them with the resources and experience neces- dates. “Patheon’s openness to the integration of these types of
sary to build upon their existing manufacturing capacity to success- technologies into their network maximizes Pacira’s ability to sup-
fully manufacture and globally commercialize EXPAREL. After ply EXPAREL to patients, as well as our future pipeline,” says
vetting other prospects, Pacira decided on Patheon. Kaczmarek Kaczmarek. “By having these strategic discussions and agree-
says what was particularly appealing about Patheon is that the re- ments, it minimizes the cost of manufacturing for both companies
lationship between the two companies is not a typical CDMO/spon- and it maximizes the ability to provide patients the quality drug
sor relationship. “I’m not just handing the drug product over and products in a very timely manner. Ultimately, that’s what we want to
then hoping they manufacture and provide the supply to me,” he be able to do, and both companies are coming together to make
explains. “We’re working together to transfer a complex manufac- that possible.” ■

42 www.patheon.com ● doingbusiness@patheon.com
Client
Success
Stories

Fighting the
Opioid Epidemic:
How Grünenthal’s
Abuse-Deterrent
Technology
Contributes
Title Goes Here

Prescription opioid products are an important component in the have the ability to inhibit this progression, thereby preventing many
armamentarium of modern pain treatment. While the wider use of users from moving to the next phase of addiction, which is com-
opioids has led to many benefits for patients in pain, it also has led monly heroin. The issue of heroin addiction is also a growing issue,
to higher incidences of misuse, abuse, and development of opioid as the number of overdose deaths from this cheaper, more acces-
addiction. Extended release formulations of opioids, in particular, sible drug has quadrupled since 2010.4
have become a major factor in prescription drug overdose deaths
in the U.S. over the last 20 years. According to the Centers for Dis- Finally, the growth of prescription abuse and the progression of the
ease Control and Prevention, more than 183,000 people died in the addiction have impacted the medical community as well, creating
U.S. between 1999 and 2015 from overdoses related to prescrip- negative reputations for both pharmaceutical companies and the
physicians prescribing opioids. This black cloud over the com-
tion opioids.1 It is estimated that U.S. hospital emergency rooms
pound class of opioids may become so damaging that, in the future,
see more than 1,000 people each day as a result of prescription
eligible patients might not have access to this valuable treatment
opioid abuse, and 91 Americans die daily from an overdose related
option for pain. In fact, issues with availability have already begun
to prescription opioids and/or heroin.2
to negatively impact many patients’ access to pain relief medica-
As part of the FDA’s efforts to address this growing issue, the agency tions due to some state-level restrictions5 and rescheduling for the
put an Opioid Action Plan into effect in 2011 to “reduce opioid abuse, hydrocodone combination products.6
dependence, and overdose in the United States.”3 Part of this plan in-
In the 2000s, Grünenthal, an international pharmaceutical compa-
cluded a call to pharmaceutical manufacturers to develop and expand
ny with a fully integrated R&D, took notice of the growing rate of
access to abuse-deterrent formulations (ADFs) in order to discourage
opioid abuse. The issue concerned the Grünenthal team not just
abuse among patients and promote innovation in the industry.

Among other benefits of ADFs, these formulations can create a 1. Centers for Disease Control and Prevention, Prescription Opioid Overdose Data
— https://www.cdc.gov/drugoverdose/data/overdose.Html
hurdle to abuse progression for recreational abusers. This is be- 2. Centers for Disease Control and Prevention, Understanding the Epidemic —
cause patients often start abusing prescription drugs orally, either https://www.cdc.gov/drugoverdose/epidemic/
by chewing or swallowing them whole, but during the course of 3. FDA, Fact Sheet — FDA Opioids Action Plan — https://www.fda.gov/
newsevents/newsroom/factsheets/ucm484714.htm
their dependency, eventually switch to non-oral routes. This typical- 4. Centers for Disease Control and Prevention, Heroin Overdose Data — https://
ly begins with intranasal insufflation due to the nasal cavity being a www.cdc.gov/drugoverdose/data/heroin.html
thin, well-vascularized tissue, which allows faster delivery through 5. ACP Internist, States aim to limit opioid prescriptions — https://acpinternist.org/
archives/2016/10/laws.htm
a direct route to the bloodstream. Once a user develops a certain 6. FDA Blog, Rescheduling prescription hydrocodone combination drug products:
tolerance to the intranasal route he or she then switches to intrave- An important step toward controlling misuse and abuse — https://blogs.fda.gov/
fdavoice/index.php/2014/10/re-scheduling-prescriptionhydrocodone-combination-
nous injection, which can lead to the sharing of needles. ADFs drug-products-an-importantstep-toward-controllingmisuse-and-abuse/

www.patheon.com ● doingbusiness@patheon.com 43
from a perspective of patient safety, but they also did not want to stream operations require a well-trained and educated manufactur-
see legitimate patients prevented from receiving necessary medi- ing staff to run the process reliably. Finally, Grünenthal’s ADF cus-
cation. The company set out to create an ADF with a technology tomers are currently all U.S.-based companies, which called for a
that creates a physico-chemical barrier to abuse and, as a result, development and manufacturing site in the U.S., to simplify the han-
safeguards patients from the battle of addiction. dling of scheduled drugs. To achieve these goals, Grünenthal sought
out a partner with the expertise, capacity, and location that could
accommodate its needs and drive the success of INTAC in the U.S.
A formulation against manipulation
Grünenthal’s abuse-deterrent formulation, INTAC®, guards against Making the cut
manipulation techniques often utilized by prescription abusers.
These usually involve pulverization and, sometimes, subsequent In addition to the practical needs mentioned above, Grünenthal
dissolution, of the tablets. Grünenthal developed a proprietary pro- sought a CDMO with a solid reputation in the pharmaceutical in-
cess using hot melt extrusion to apply heat and pressure to create dustry for its expertise, reliability, and cost, as well as a willingness
a crush-resistant matrix. The resulting extrudate has the resistance to embrace the challenge of implementing a specialized technolo-
and gelling properties needed to make INTAC’s barrier successful. gy. Grünenthal ultimately selected Patheon, which was an
The process guards against manipulation while still maintaining the especially appealing option due to the company’s condominium
clinical benefit of the standard form for the patient. The result is a manufacturing model. The condo model is one of six adaptable
formulation that deters abusers but does not require the patient to manufacturing arrangements Patheon offers its clients. This highly
take additional ingredients that do not add to their therapy or may customizable manufacturing solution provides sponsors with a va-
precipitate withdrawal. riety of flexible and scalable approaches to create the optimal solu-
tion to fit their program’s unique needs.
By making the formulation resistant against crushing, Grünenthal
impedes intranasal abuse, because the tablet cannot be turned For Grünenthal, Patheon and its condo model addressed each of
into a fine powder. This also prevents intravenous abuse, as abus- INTAC’s biggest manufacturing challenges. Both companies
ers usually first disintegrate the tablet and then dissolve the powder worked together to plan a layout for the manufacturing suite dedi-
cated to the needs of INTAC. “Because of the flexibility offered by
in small amounts of liquid, which they subsequently inject. With
Patheon, we were able to optimally set up the equipment in the
INTAC, such attempts lead to the creation of a viscous mass that is
INTAC suite without any restrictions,” explains Siegfried Ebner,
difficult to draw into a syringe. While it is impossible to completely
Head of the Innovation Unit Devices and Technologies at
prevent abuse, research by the CDC indicates barriers like these
Grünenthal. “In addition, operators and engineers traveled to our
can reduce the progression of abuse from oral to non-oral routes.7
Aachen headquarters for training on the INTAC equipment long
The INTAC platform addresses various release profiles, from ex- before it was installed in the Patheon suite. Patheon’s Cincinnati
tended release (ER) to immediate release (IR) and a wide variety of location addressed another major need for us, as it brings the IN-
mixed ER/IR profile (also known as modified release, or MR). Be- TAC technology infinitely closer to U.S. customers, eliminating the
sides the release profiles, the technology also allows formulating need for further transfers. As a result of working intensively togeth-
various substance classes. Specifically, there has been a positive er from the beginning, Patheon’s staff was ultimately able to run our
experience when creating INTAC-based formulations of prescrip- equipment and processes with complete success.” While distance
tion stimulants. These drugs are also being abused for recreational can be a hurdle in any global relationship, the companies were able
purposes via snorting and injection. to overcome the 6-hour time difference between Aachen and Cin-
cinnati with regular teleconferences as well as for any issues that
required special attention. Meeting face-to-face on a regular basis
From extrusion to the tablet also proved beneficial as each team believes this strengthens the
relationship of all involved.
While Grünenthal’s core manufacturing process for extrusion is
rather straightforward, it is the combination of certain excipients like Grünenthal further continues its development of the INTAC tech-
polyethylene oxide (PEO) that, under hot melt extrusion, lead to the nology as a way to help fight against the growth of opioid abuse and
unique properties of INTAC formulation. The proprietary down- protect patients from this deadly epidemic. Looking forward, the
stream process uses extrusion, cutting, and cooling for the formu- company believes its technology is suitable to raise the hurdle to
lation of an abuse-deterrent final dosage form that can be further abuse of other compounds, that are prone to misuse and abuse.
Projects with these compound classes are under development,
processed into tablets. The gelling properties of the excipients
and research is also being done to investigate the suitability of
used in the formulation are maintained, thereby sustaining the bar-
Grünenthal’s technology to impede conversion from pseudoephed-
rier to injection.
rine to crystal meth. With a strong contender for widespread abuse
During development, manufacturing of INTAC presented several prevention and a trusted partner on its side, Grünenthal stands
challenges. First, although the extrusion technology used is well strong as an innovator in pharma as well as an advocate for the
known in the industry, it requires specific expertise to utilize the pro- health and safety of today’s patients. ■
cess optimally. Second, specialized equipment is required to estab-
lish a manufacturing process that can run almost continuously from 7. J. Ziebell, presentation at Nat Rx Abuse & Heroin Summit 2016 - https://www.fda.
gov/downloads/Drugs/NewsEvents/UCM527899.pdf
extrusion to the solid-dose form. Third, the nonstandard down-

44 www.patheon.com ● doingbusiness@patheon.com
Brilliant
discoveries.
Delivered.

We are Patheon, and we bring to


bear 40 years of experience and
expertise, from development to
manufacturing. We also bring global
reach. An industry reputation for
being right on time, the first time.
Supply chain solutions designed
to simplify complexity and speed
up the process. And a passionate
belief that together we can make
the world a healthier place.
BIOLOGIC DRUG SUBSTANCE API DRUG SUBSTANCE

Ahmedabad, India
Allentown, PA, USA
Basel, Switzerland
Beijing, China
Bend, OR, USA
Bourgoin, France
Brisbane, Australia ●
Buenos Aires, Argentina
Cincinnati, OH, USA
Ferentino, Italy
Florence, SC (West), USA ●
Florence, SC (East), USA ●
Greenville, SC, USA ●
Greenville, NC, USA
Groningen, Netherlands ●
High Point, NC, USA
Horsham, United Kingdom
Indianapolis, IN, USA
Linz, Austria ●
Manatí, Puerto Rico
Mexico City, Mexico
Milton Park, United Kingdom
Monza, Italy
Moscow, Russia
Mt. Prospect, IL, USA
Pretoria, South Africa
Princeton, NJ, USA ●
Regensburg, Germany ●
São Paulo, Brazil
Seoul, Korea
Singapore
St. Louis, MO, USA ●
Suzhou, China
Swindon, United Kingdom
Tilburg, Netherlands
Tokyo, Japan
Toronto, Canada
Weil am Rhein, Germany
Whitby, Canada
OUR INTEGRATED GLOBAL NETWORK
PHARMACEUTICAL CLINICAL TRIALS SUPPLY
DRUG PRODUCT SERVICES
DEVELOPMENT SERVICES CHAIN SERVICES





● ●


● ●
● ●

● ●

● ●











● ●

● ●

● ●
Patheon
4815 Emperor Blvd, Suite 300
Durham NC 27703-8470 USA
P: +1 919 226 3200
F: +1 919 474 2269
www.patheon.com

Patheon
Kingfisher Drive
Covingham, Swindon
Wiltshire SN3 5BZ UK
P: +44 1793 524411
F: +44 1793 487053
www.patheon.com

Patheon
7F Wakamatsu Building, 3-3-6
Nihonbashi Hon-cho, Chuo-ku,
Tokyo 103-0023
Japan
P: +81 3 6202 7666
F: +81 3 6202 7676
www.patheon.jp

Potrebbero piacerti anche