Download as pdf or txt
Download as pdf or txt
You are on page 1of 28

APIs: Chemistry to

Commercialization

1
Table of Contents

1
Introduction

Chemistry & Innovation


Synthetic Route Scouting: Factors to Improve API Manufacturing
6 Properties of API Synthesis Affecting Yield, Delivery Date & Purity

2-13
API Manufacturing of Deuterated Molecules
Peptide Synthesis: Solution Phase, Solid Phase or Hybrid?
Purification of Peptides Using Surrogate Stationary Phases on Reversed-Phase Columns
Disrupting Peptide Aggregation

Manufacturing & Commercialization

14-19
Common Challenges of Scaling Up an API
Contract API Manufacturing: From Micro-batches to Multi-Tons
API Micronization Techniques for Use in Injectables and Medical Devices

Quality & Regulatory


Genotoxic Impurities: Increasing Vigilance, But Still Some Uncertainty
Leveraging QbD for API Scale Up
How to Strengthen Your API Supply Chain Management and Sourcing
20-25
Introduction
Small molecule APIs and peptides are a story
of continuous innovation – from chemistry
through to commercialization.
Today – as API development and
commercialization has grown more complex
– the industry is increasingly demanding
faster, simpler, cleaner and more cost-
effective manufacturing techniques to
address growing drug pricing pressures.
For 35 years, from our first API sale in the
mid-1980s to our 32nd successful regulatory
inspection in 2019, Neuland has invested in and expanded into
multiple facilities, broadened our capabilities and built a robust
track record of global regulatory excellence. Our customers are
at the core of everything we do.
Neuland’s expertise in synthetic chemistry and process development provide our customers and
partners with confidence that their drug intermediates will meet or exceed standards – on time,
every time. We have become a trusted generic and NCE API manufacturing partner & supplier to
some of the biggest names in global pharma.
We currently produce 75+ APIs for the customers across 80+ countries in 10 diverse therapeutic
segments.
This collection of articles discusses some of the key factors driving the pharmaceutical API
industry – from Quality by Design to groundbreaking chemistry advances in the fields of peptides,
deuterated molecules and process optimization.
We hope you find these insights helpful! Please visit our website or contact us to learn more about
Neuland’s capabilities.

Saharsh Davuluri
Joint Managing Director
Neuland Laboratories Ltd.

1
Chemistry
& Innovation

In this section, we present a In 6 Properties of API Synthesis Due to their efficacy and safety,
collection of articles focused on Affecting Yield, Delivery Date & Purity peptide-based therapeutics
a number of pharmaceutical API we discuss some of the key synthesis are increasingly viewed as an
chemistry innovations driving change attributes which our team at extraordinary opportunity for
in the drug industry. Neuland have found play a role in the pharmaceutical companies.
successful manufacture of an API. In Peptide Synthesis: Solution
At Neuland Labs, our expertise is
Phase, Solid Phase or Hybrid?,
often called on to provide custom Deuterated compounds – in which
Disrupting Peptide Aggregation,
synthesis and route scouting. drug molecules are modified with
and Purification of Peptides Using
Demand for these services deuterium to extend the drug’s half-
Surrogate Stationary Phases on
has continued to grow as more life – continue to show promise in
Reversed-Phase Columns, we share
companies realize the cost, efficiency potentially boosting the bioavailability
some fundamental peptide API
and safety benefits of process and safety of some drugs. API
principles, along with successful
optimization. Synthetic Route Manufacturing of Deuterated
Neuland innovations designed for
Scouting: Factors to Improve API Molecules highlights some of the
feasible, cost-effective large-scale
Manufacturing explores some of the issues involved in this emerging
manufacturing.
top things to consider when devising specialized field.
new routes.

2
Synthetic Route Scouting: Factors to
Improve API Manufacturing
Synthetic route selection is a crucial Ask the Right Questions to Keys to Getting Route
element in API manufacturing. While Improve Alternate Routes Scouting Right
the requirements of the synthetic
process of a drug will naturally • When considering a route When considering options, keep
evolve during its life cycle, scouting change, the first question to these final points in mind to achieve
alternate routes early in process ask is why. By changing the success:
development provides many benefits. route, what clear benefits can be
• An alternative route should use a
Alternate routes have the potential to gained in terms of cost, time and
strategically inexpensive starting
help: availability?
material. Using an intermediate
• The next point to consider is the
• Improve scalability from an existing process is ideal.
availability of raw materials. Are
• Reduce chemical or reagent • The process used should be
they maximized by your current
usage and waste production robust and require minimal
process, or could an alternate
• Decrease processing times purification. Stages should
process improve on it? Look at
• Improve quality and safety profiles be telescoped for maximum
the volume of the product in the
• Reduce the number of processing efficiency.
market. The higher the volume
steps or overall complexity • The new route should offer
you need to produce, the more
high “atom economy,” creating
Essential Drivers of benefit you gain from improving
minimal waste via a greener
API Route Scouting the route.
process.
• Does the manufacturer you
When developing a new synthetic API • And finally, the developed route
workwith conduct company
route, we look for a route that: must not infringe on any current
product profile matching?
patents.
• Is cost-effective This can be very helpful when
• Has the same quality or seeking to make a change. When Keeping these factors in mind during
greater quality than the previously examining the cost benefit, does the research phase should allow the
agreed upon route it extend to the manufacturer as creation of a new synthetic route
• Provides reasonable time to well as to you? that meets expectations and is
market • Is the process feasible in the sustainable across the drug lifecycle.
manufacturing plants you’ve
While meeting the three criteria selected?
above would yield the most benefit, • Lastly, what alternate green
focusing on even one or two of reagents could be used for
these criteria can provide significant long-term sustainability of the
process improvements. For niche product?
products, most companies seek
to reduce cost by 2-5% through Today, the most innovative routes
alternate routes; for generics, much use the least resources possible and
greater savings are sought. minimize impact on the environment.
In addition to cost, other factors to
consider are batch sizes, throughput
of the product, lead time and
reducing batch cycle. Shortening the
route is frequently a goal, as this one
change can singlehandedly decrease
cost, time, waste and regulatory
constraints.
6 Properties of API Synthesis Affecting
Yield, Delivery Date & Purity

The ultimate goal is synthesizing an Parallel synthesis reduces the Synthesizing each distinctly charged
API product with high yields, ease number of steps that are dependent part separately as intermediates may
of isolation from impurities, and no on receiving an intermediate by simplify the isolation strategies and
off-target hazardous impurities. Final first preparing each part of the synthetic steps. Intermediates can
products with these characteristics API molecule. The intermediate react at the correct position on a core
are more likely to pass stringent FDA molecules are then reacted in structure or intermediates can be
and EU GMP requirements. a predesigned sequence that attached to each other at the correct
synthesizes the complete product. position until the final product is
However, there are a number of
produced.
properties of API synthesis that can The steps leading to the intermediate
affect the yield, purity, and delivery molecules dictate the structure
date of a final product. of the impurities. The selection of
the intermediate molecules and
1 Type of synthesis directly their route steps can minimize the
affects yield, delivery date, possibility of off-target synthesis of
and purity. hazardous contaminants.

Linear synthesis of the API The intermediates may differ in their


consistently modifies each step in a charge properties, which allow the
sequence. In a multi-step synthesis intermediates to be easily isolated
with yields of 80% at each step, the from impurities with a single column
overall yield drops by 20% for each pass. Often complex molecules have
individual step. Thus, the total yield mostly positive or mostly negative or
of a 6-step process is approximately mostly neutral (hydrophobic) charges
26.2% and yield for an 8-step process in different regions.
is 16.8% (Table 1).
The unreacted intermediates, B,
C, D, E, F, G and H in our example
below need to be purified from
intermediate(s) or finished products
of each reaction.

Table 1. Comparison of yields and timing of steps between linear and


parallel routes of synthesis

Linear synthesis Parallel synthesis


Route Steps 80% yield for each step Route steps 80% yield for each step
1 A→B 80.0% 1 A→B 80.0%
2 B→C 64.0% 1 C’ → D’ 80.0%
3 C→D 51.2% 1 E’ → F’ 80.0%
4 D→E 41.0% 1 G’ → H’ 80.0%
5 E→F 32.8% 2 B + D’→D 64.0%
6 F→G 26.2% 2 F’ + H’→J’ 64.0%
7 G→H 21.0% 3 D + J’ → J 51.2%
8 H→J 16.8%
Total yield 16.8% 51.2%

4
2. Propinquity The manufacturing of some
pharmaceuticals results in isomers.
In Table 1, the Propinquity or number Isomers are like a right wrist and
of steps in the synthesis route a left wrist—the bending of the
directly affects the yield. Since each wrist and its ability to rotate are
step has its unreacted components, critical for it to function optimally …
the number of steps also affects similar to complex molecules. If an
the number of isolation steps and intermediate step yields isomers, 6. Minimum isolation steps in
impurities. The number of steps the active isomer may be more easily situ
that can be done simultaneously isolated in some cases.
by different chemists can reduce Typically, synthetic routes are
delivery time, whereas downstream Parallel synthesis helps reduce designed to utilize single or two
steps cannot be started until the the number of potential isomeric isolation steps for the intermediates.
intermediate(s) are done and variants. For example, Labetalol has By making intermediates in a parallel
purified. four isomers in its racemic mixture. manner, certain columns such as
Two are relatively inactive. The potent specific anion or cation columns
Generally, manufacturers tend to alpha blocking activity comes from can be used more easily for their
prefer developing synthetic processes the (S,R) isomer component and the isolation. Due to efficient stepwise
under non-GMP regulations, while potent beta blocking activity derives isolation of intermediates, the
manufacturing APIs under current from its (R,R) isomer. isolation of the final product is less
Good Manufacturing Practices cumbersome and generally has a
(cGMP). This method – which aims 4. Cost-efficient synthesis very high purity.
to reduce the number of steps – is Synthetic route planning is critical
preferred by manufacturers for three to maximizing yields. As shown in
reasons: Table 1, parallel synthetic strategies
• Reduces costly GMP manufacture overlap the scheduling of the
• Reduces reporting of process synthesis of intermediates, and can
variation and optimization reduce cycle time, and minimize the
• Increases flexibility of process and number of downstream processing
sourcing steps.
Regulators tend to request more 5. Carryover of impurities into
steps in the GMP manufacturing drug substance
process for three main reasons: Synthetic routes that minimize or
• Minimizes quality risks eliminate any potential off-target
• Controls impurities from starting synthesis of hazardous or unknown
material and subsequent steps impurities should be utilized. Green
and to ensure identity and purity chemistry has become increasingly
of drug substance important, so routes should be
• Purges related impurities with planned to minimize effluent
multiple purification steps. generation. This can reduce the time
or even the need to concentrate the
3. Complexity of structure intermediates.
Generally, a more complex structure
usually involves more synthetic steps
for the API.

Manufacturer tendency: Less steps under cGMP

Non-GMP GMP

SM1+SM2 Int-1
Int-3
Int-4 DS DS-Salt
(RSM1)

Int-5
SM3+SM4 Int-2
RSM2

Legends: Regulator tendency: More steps under cGMP


SM : Starting Material
Int : Intermediate
RSM : Regulatory Starting Material
DS : Drug substance

5
API Manufacturing of Deuterated Molecules
Deuterated compounds – in which The deuterated compound market The chart below represents
a drug molecule’s carbon-hydrogen has attracted many new companies indications and clinical status
bond is replaced with a carbon looking to develop and patent for deuterated candidates from a
deuterium bond to extend the deuterated versions of various number of companies in the space.
drug’s half-life – continue to show existing, non-deuterated therapeutic
promise in potentially boosting the compounds. This is known as the
bioavailability and safety of some “Deuterium Switch.”
drugs.
Drug Name Active Indications Highest Status
deuterated D-serine (oral, Schizophrenia Phase 1 Clinical
schizophrenia), CoNCERT
Pharmaceuticals
PXL-065 Adrenoleukodystrophy; Adrenomyeloneuropathy; Phase 1 Clinical
Non-alcoholic steatohepatitis
ALK-001 Age related macular degeneration; Cone dystrophy; Phase 2 Clinical
Retinitis pigmentosa; Stargardt disease
deuterated S-lisofylline Necrosis; Radiation sickness Phase 2 Clinical
deudextromethorphan hydrobromide Agitation; Intermittent explosive disorder; Phase 3 Clinical
+ ultra-low dose quinidine sulfate Schizophrenia
11,11-di-deutero-ethyl linoleate, Brain disease; Friedreich ataxia; Huntingtons chorea; Phase 3 Clinical
Retrotope Motor neurone disease; Neurodegenerative disease;
Progressive supranuclear palsy; Tay Sachs disease
deutivacaftor Cystic fibrosis Phase 2 Clinical
deuterium-modified ruxolitinib Alopecia areata Phase 2 Clinical
analog (oral, alopecia areata),
CoNCERT Pharmaceuticals
CTP-730 Inflammatory disease Phase 1 Clinical
HC-1119 Breast tumor; Hormone refractory prostate cancer; Phase 3 Clinical
Prostate tumor
JZP-386 Narcolepsy Phase 1 Clinical

What is a Deuterated Drug?


A deuterated drug is a small
molecule with medicinal activity. It
is made by replacing one or more of
the hydrogen atoms contained in the
drug molecule with deuterium – a
hydrogen isotope whose nucleus
contains one neutron and one proton.
As deuterium and hydrogen have
nearly the same physical properties,
deuterium substitution is the
smallest structural change that can
be made to a molecule.
To Deuterate or Not to
Deuterate – That’s the
Regulatory Question
Developers of deuterium switch
compounds must show significant
clinical benefits over existing non-
deuterated versions to justify why
they should replace existing or less
expensive therapies. Such a switch
can:

6
• Take advantage of the clinical
knowledge concerning the
non-deuterated version of the
compound.
• Benefit from new patent
protections.
• Result in improved therapies and
patient outcomes.
Most large pharmaceutical
companies today also claim
deuterated versions of new
molecules in their patent
applications.
Benefits of Deuterated
Versions of Drugs
Deuterated versions of existing
drugs can benefit from improved
pharmacokinetic or toxicological
properties. Because of the kinetic
isotope effect, which is the change
in rate of a chemical reaction when
one of the atoms in the reactants is
substituted with one of the isotopes,
drugs that contain deuterium may
have significantly lower metabolism
rates. As the C-D bond is ten times
stronger than the C-H bond, it is
much more resistant to chemical or
enzymatic cleavage and the difficulty
of breaking the bond can decrease
the rate of metabolism. Manufacturing Deuterium the drug was found to reduce the
Exchanged APIs disease symptoms and the frequency
Lower metabolism rates give of administration, and it is currently
deuterated drugs a longer half- With our expertise in deuteration being considered for approval by the
life, lengthening the timeline for technology, Neuland Labs uses a FDA.
elimination from the body. This synthetic approach where deuterium-
enriched material is combined with Recently, another investigational
reduced metabolism can extend a
the drug to produce deuterated new drug targeting
drug’s desired effects, diminish its
drugs. Another approach, called an nonalcoholic steatohepatitis
undesirable effects, and allow less
exchange approach, uses a catalyst & adrenomyeloneuropathy (a
frequent dosing. The replacement
to produce a deuterated molecule. deuterium-stabilized [R]-enantiomer
may also lower toxicity by reducing
of pioglitazone) completed FDA
toxic metabolite formation. The most popular process for review and appears headed towards a
A major potential advantage of sourcing deuterium for drugs is Phase 1 study.
deuterated compounds is the extracting D2O from regular water via
the Girdler sulfide (also known as the Growing Opportunity for
possibility of faster, more efficient,
Geib-Spevack) process, which uses a Deuterated Drugs
less costly clinical trials, because
of the extensive testing the non- temperature difference and hydrogen The current market value of
deuterated versions have previously sulfide to enrich deuterium in water companies specializing in this
undergone. The main reasons by up to 20%. technology suggests that the value
compounds fail during clinical Deuterated Molecules of “deuterium switching” could be
trials are lack of efficacy, poor Advance in Clinical Trials more than a $1 billion, and that the
pharmacokinetics or toxicity. With greatest discoveries in the field have
While deuteration has been around yet to occur.
deuterated drugs, efficacy is not in
for decades, most deuterium
question – allowing the research
chemistry efforts are currently in the
to focus on pharmacokinetics and
pre-formulation stage.
toxicity.
Those deuterated compounds
Deuterated versions of drugs might
that have advanced are generally
also be able to obtain FDA approval
performing well in clinical trials.
via a 505(b)(2) NDA filing, a faster,
In July of 2016, a deuterated drug
less expensive route.
reached Phase III testing for the first
time, in a study to treat Huntington’s
disease. Known as deutetrabenazine,

7
Peptide Synthesis: Solution Phase,
Solid Phase or Hybrid?
Peptides are a complex drug Choosing the Right Synthesis • The ultimate commercial launch
class, and have historically proven Technique for Your Peptide quantities & batch sizes that will
challenging from a manufacturing API be required for manufacturing
standpoint. They are, however,
Peptide synthesis has roots dating Peptides – Different
experiencing a renaissance due to
back more than a century. Given its Production Methods
improvements in peptide synthesis,
the development of high-throughput age, it may seem strange that peptide Peptides are produced using one
approaches and various innovations synthesis is currently undergoing a of three synthesis methods: liquid
to overcome some of their traditional revolution…but it is. A new generation phase, solid phase or a hybrid
limitations, such as stability and half- of therapeutics, diagnostics and approach. Each has its advantages
life. These advances are expected to research tools are emerging on the and disadvantages.
drive the peptide drug market to over healthcare scene – a by-product of
the industry’s turn towards biologics • Liquid Phase: 15 and Fewer
$48 billion by 2025.
and a greater understanding of the Amino Acids
There are three main strategies benefits of peptides. Solution phase synthesis (commonly
for synthesizing peptide active
The decision regarding which referred to as ‘liquid phase’) is
pharmaceutical ingredients (APIs):
production technique to use is driven regarded as the traditional approach
• Solid Phase by three pivotal factors: to peptide production. Among its
• Liquid Phase benefits, solution phase synthesis
• Hybrid Technology • The size of peptide (meaning the delivers better economies of scale.
number of amino acids) The technique is much more scalable
• The quantities needed at the and produces large quantities of
current stage of development

8
high-quality peptides at a lower cost construct a 40-amino acid peptide,
point than solid phase or hybrid small peptides of 5 to 8 amino acid
methods. Solution phase is not well- segments would be produced using
suited to the production of larger solid phase methods on resins, and
peptides, but it is an ideal strategy then the segment condensations
for peptides containing less than 15 would occur in solution to construct
amino acids and when commercial the full peptide sequence.
requirements range from 10+
Today, a hybrid peptide synthesis
kilograms to tons.
strategy is generally chosen for Liquid Phase & Hybrid
• Solid Phase: 25 Amino Acids peptides that are greater than Synthesis for Improved API
& Up 25 amino acids in length, and Substance Profiles
commercial requirements range from
In solid phase synthesis, the peptide Whenever possible, transitioning
10 to 200 kilograms. One notable
is constructed on resins (e.g., from solid phase synthesis to a
success from hybrid synthesis was
polystyrene, polyacrylamide, PEG). hybrid – and ultimately liquid phase
the pioneering work on Fuzeon, an
The key advantage with solid phase – methodology is recommended due
approved 36 amino acid therapeutic
is the ability to synthesize peptides to the successive improvements in
for HIV-1.
which don’t lend themselves to purification. With each change in
bacterial expression using solution Choosing the Right Production strategy – from solid to hybrid, and
phase techniques. Method from hybrid to liquid – significant
One of the major challenges facing The choice of which method to use improvements in the substance
solid phase synthesis, however, is is generally guided by a number of profile are observed with peptide
yield – as the size of the peptide factors. A key factor is the desired APIs.
increases, yields typically decrease chiral purity/integrity of the peptide. Peptide APIs produced with a solid
due to the challenge of removing Pseudoprolines can be exploited for phase approach contain inherent
closely related impurities from the racemization-free condensations of deletion- and insertion-related
product. And while some molecules segments terminating in a serine substances. These substances
don’t lend themselves to bacterial or threonine residue. This strategy decrease progressively with the
synthesis, others aren’t well-adapted maintains purity through the various transition to hybrid or liquid phase
to solid phase synthesis – either segment synthesis steps – especially techniques. As peptide production
because of the inherent aggregation with longer (or more difficult to progresses across these 3 distinct
encountered during the assembly produce) peptides. methods, the intervening purification
of longer peptides, or due to the steps increase – leading to fewer
Factors which drive the choice of
chemicals used to remove them from unwanted substances in the peptides.
synthesis method include:
the resin – which can damage the
peptide. Production costs also tend to 1 The number of amino acids in the
be substantially higher in solid phase peptide
synthesis.
2 The scale of production (gram
Conventional wisdom suggests using or sub-gram up through kilogram
a solid phase approach for peptides quantities)
containing greater than 25 amino
3 The timeframe required for the
acids for commercial quantities
peptide
in the 1-10 kg range. Solid phase
peptide synthesis utilizes an excess
of protected amino acids to ensure as
close to a 100% complete reaction as
possible.
When larger quantities are needed,
this can add considerable cost to
the synthesis. However, a significant
benefit of solid phase synthesis is the
relatively shorter cycle time when
compared to liquid phase synthesis.
A secondary benefit: liquid phase
synthesis of peptides larger than 15
amino acids is labour intensive.
• Hybrid Synthesis: More Than
25 Amino Acids & Larger
Commercial Quantities
The hybrid approach brings these two
different methodologies together to
produce peptides. For example, to

9
Purification of Peptides Using
Surrogate Stationary Phases on
Reversed-Phase Columns
In recent years, peptides have been of raw materials, intermediates, and loading capacity of a given column,
acknowledged for their selectivity, APIs. Preparative RP-HPLC is used nor have they resulted in a significant
efficacy and safety. Combined with for the commercial production of increase in the amount of purified
new synthesis technologies and peptide APIs and most other complex product (output/mL of the packed
advances in peptide science, peptide APIs not suited to crystallization. column).
therapeutics have become a reality
Historically, there have been just Using RP-HPLC
– and numerous clinical trials are
two ways to increase the amount
ongoing. In science labs, reversed
of sample that could be purified
phase high performance liquid
But while peptides may be safe and in a single Prep-RP-HPLC run:
chromatography (RP-HPLC) is used
effective as drugs, they have never use a larger column (increase
to analyze, characterize, separate,
lent themselves to straightforward the stationary phase) or use
purify, and isolate small organic
commercial manufacturing – displacement chromatography.
molecules, natural products, and
especially longer-chain, complex
Advances Have Brought biologically active molecules such
peptides.
Commercial HPLC Closer as polypeptides, proteins and
In a peer-reviewed piece at nucleotides.
The technological advances in
Pharmaceutical Technology,
process HPLC instrumentation and In pharma, analytical RP-HPLC
members of the Neuland team
the bonded silica supports have made is employed specifically to
shared a newly-developed strategy
possible commercial production of release and characterize raw
to increase sample loading 7-12 fold
complex peptides such as Fuzeon, a materials, intermediates and active
compared to conventional Prep-HPLC
36-amino acid peptide, in hundreds pharmaceutical ingredients (APIs).
techniques.
of kilos quantities. Unfortunately, Likewise, preparative RP-HPLC
Reversed-phase high-performance large-scale HPLC instruments and is used to commercially produce
liquid chromatography (RP-HPLC) the associated column hardware are peptide APIs, along with most
is used throughout the pharma expensive. other complex APIs that cannot be
industry. Analytical RP-HPLC is used crystallized.
More importantly, none of these
for the release and characterization
improvements have addressed the

%
Relative
Internal Loading
Total Amount loading
diameter Type of Amount with
Source/ Type of column of capacity
Entry (cm) x stationary of crude respect
Operator Chromatography volume stationary (Standard
length phase purified to total
(ml) phase RP-HPLC
(cm) column
= 1)
volume
40 g of
1 RP-HPLC 4420 15 x 25 C-18 3.5 kg crude 0.9% 1.0
peptide
Donald A. Self 6.2 g of
Wellings, Not
2 displacement – 347 4.2 x 25 PLRP-S a 25-AA 1.8% 2.0
A Practical available
RP-HPLC peptide
Handbook of
Preparatory 120 g (6
HPLC, box-car
Enantiomers Not
3 Elsevier, UK 1964 10 x 25 Chiralpak AD injections 1.0% 1.0
separation available
(2006) (1) of 20 g each
injections)
NP-HPLC
4 196429 100 x 25 Silica 155 kg 15,500 g 7.9% 8.7
(normal phase)
Discovery
Wide Pore
TBAHS-SSP-RP- 1.4 g crude
5 19.6 1 x 25 C-18, 5μ 11 g 7.1% 7.9
HPLC leuprolide
100 A pores,
Neuland: 9.2% C
this paper Water
Symmetry
TBAHS-SSP-RP- 1.4 g crude
6 14.1 1.9 x 5 C-8, 5μ 100 11 g 9.9% 11.0
HPLC leuprolide
A pores,
11.7% C

10
How the New RP-HPLC
Method Works
The new method developed by
Neuland uses C-18/C-8 derivatized
silica, coated with a hydrophobic
quaternary ammonium salt or
quaternary phosphonium salt. It
increases 7- to 12-fold the sample
loading of the crude mixture of
organic compounds including
synthetic crude peptides. What
causes such dramatic results is the
additional surrogate stationary phase
characteristic of the C-18/C-8 bound
quaternary salt.
Preparative RP-HPLC in the elution
mode is limited by the loading
capacity of the analyte. In this mode,
however, synthetic peptides typically
have a loading capacity of 1-2 mgs
per mL of packed column volume
(viz., 0.1% to 0.2% with respect to
total column volume). Table 1 (found
on page 2 of PDF provided at web
link) shows how Prep-RP-HPLC
aided by a surrogate stationary phase Analytical RP-HPLC Profile of Leuprolide acetate obtained
performed compared to standard using the Additional Stationary Phase aided Prep-RP-HPLC
Prep-RP-HPLC using Leuprolide
as an example, and tetra-n-
butylammonium hydrogen sulphate
(TBAHS) as ASP/SSP.
Results of Neuland’s Prep
HPLC Technique
The purified product (Leuprolide)
output of the standard Prep-RP-
HPLC is 2.45 mg/mL of column
volume. In contrast, the purified
product output of the Prep-RP-HPLC
aided by a surrogate stationary phase
is 29.6 mg/mL of the column volume
Peak Results
(see Table 1, entry 2, on page 2 at
this link) and 16.3 mg/mL of column
volume (see Table 1, entry 3, on
page 2 at this link). These results
demonstrated that the process
described enables loadings of 7-12
times the capacity of conventional
Prep-RP-HPLC.
To learn more about this novel Prep- Illustrative Example
RP-HPLC method, view the complete
PDF here. The C-18/C-8 reversed column is and loaded on the column. After
equilibrated with 5 to 10 column the loading is complete, the column
volumes (VCS) of 5 to 10% aqueous is equilibrated with with 2 VCS of
acetronitrile containing 10 mM buffer A. Next, the gradient elution
TBAHS. The pH of the starting process is started. The buffer B is
buffer was not adjusted, and was usually 300 mM to 500 mM TBAHS
about 1.95 (It is important to keep in 5 to 10% aqueous acetronitrile.
the concentration of acetronitrile A linear gradient of 0%B to 100%
lower than the concentration Buffer B over 10 VCS is applied.
needed to elute the product on When the product of interest (API)
an analytical HPLC column). The is about to elute, a gradient hold
crude compound to be purified may be applied until all the API has
was dissolved in starting buffer A eluted from the column.
or aqueous TFA or aqueous HOAc

11
Disrupting Peptide Aggregation

The aggregation of peptide chains Fmoc & Orthogonal • Fmoc-pseudoproline


caused by intramolecular hydrogen Approaches to Peptide dipeptides
bonds is a common challenge with Synthesis Fmoc-pseudoproline dipeptides
longer or more complex peptides. It
One of Fmoc’s greatest advantages are the most common approach for
can result in slower and incomplete
is its ability to work well with other disrupting aggregation. The position
coupling reactions and incomplete
protecting groups (e.g., Boc) – of “Serine” or “Threonine” in the
deprotection of the Na-amino
allowing for an orthogonal approach sequence of the peptide is identified.
protecting group – meaning a
– a common strategy in organic Instead of coupling the “Serine” as
modified or damaged peptide.
peptide synthesis. a single residue (Fmoc-Ser-OH), the
There are a number of steps taken to dipeptide sequence that includes
prevent aggregation during peptide Common Fmoc Methods the next amino acid (Xxx) in the
synthesis, including cleavage and for Disrupting Peptide sequence after serine/threonine is
deprotection. The most commonly Aggregation coupled as Fmoc- Xxx-Ser/Thr-OH
used – and mildest – method is Advances in peptide synthesis pseudoproline dipeptide. Using this
Fmoc – the removal of the Fmoc methods and ready availability of approach, peptides containing over
group to expose the α-amino group. reagents that disrupt intramolecular 120 amino acids can (and have been)
In addition to cleaving under very hydrogen bonds have made complex created.
mild conditions, it is (typically, though syntheses much more practical.
not always) stable under acidic There are three Fmoc strategies for
conditions as well. disrupting aggregation. The decision
to use each one is directly dependent
on the type of building block being
used.

Intermediates Analytical and


Generic Peptide API
Research and Peptide Manufacturing Services Regulatory
Manufacturing
Fragments Support
Aid in drug design Novel Routes for Pre-GMP and scale up Analytical cGMP Manufacturing
and Analog Synthesis supply of Clinical Trial Method of Peptide APIs with
Synthesis Material Development Regulatory Support
and Validation
Custom Process Supply of Scale up from Lab to Pilot Impurity
Development Protected and Commercialisation Profiling,
Amino Acids and Validation
Building Blocks
Analytical
Reference
Standards
Stability
Studies
Filing DMFs
NEULAND’S PEPTIDE WORLD
From Building Blocks to Commercial Production to Peptide APIs
Core Expertise:
Ÿ Solution Phase Synthesis
Ÿ Solid Phase Synthesis
Ÿ Hybrid Technology for Complex Peptides

12
• Fmoc-isoacyl dipeptides
Fmoc-isoacyl dipeptides are proving themselves to be a remarkable tool for
enhancing synthetic efficiency in the Fmoc/ tbutyl based solid phase strategy.
Substitution of Xxx-Ser/Thr in a peptide sequence with the corresponding
isoacyl dipeptide results in the formation of a depsipeptide analog of the
native sequence. This modification results in the disruption of aggregation in a
manner similar to pseudoproline or N-Dmb/Hmb-residue.
• Fmoc-(Dmb)Gly-OH and analogs
The use of Fmoc- Asp(OtBu)-(Dmb)Gly-OH in sequences containing –Asp-
Gly- has been shown to suppress formation of b-Aspartyl peptides. Fmoc-
Gly-(Dmb)Gly-OH is potentially useful in the synthesis of peptides containing
multiple glycines.

13
Manufacturing
& Commercialization

In this section, Neuland’s team API particle size plays a fundamental


highlights some of the challenges role in the properties of drugs,
and techniques common to the scale- directly impacting solubility, efficacy
up and manufacture of drug APIs. and other characteristics. Advances
in particle reduction techniques
From reactions, to crystallization to
allow for improvements in API
particle size, Common Challenges
performance. In API Micronization
Scaling Up an API discusses some
Techniques for Use in Injectables and
of the specific challenges drug
Medical Devices, the Neuland team
companies face when translating
examines top techniques used today
bench processes to commercial
by pharmaceutical manufacturers.
scale.
Contract API Manufacturing: From
Micro-Batches to Multi-Tons
explores some of the key challenges
which can emerge at larger scales of
API production.

14
Common 2. Safety Data Generation

Challenges of Runaway reactions are a challenge


during scale-up. Generating safety
Scaling Up an API data at lab scale can potentially
help in designing the hardware
While linear drug scale-up sounds for pilot or commercial scale
great on paper, the reality is usually batches – leading to inherently
much different. In fact, scaling up an safer process technology. This
API can be challenging even under typically involves reactions hazards
the best circumstances. and thermal hazards studies. including the super saturation driving
These are performed using reactor force and its impact of residual
Most scale-up challenges arise from
calorimeters, Thermal Screening solvent.
the non-linear nature of shifting
units (TSu), or Rapid Screening
from the bench to the manufacturing 5. Particle Size
Devices (RSD) to monitor:
plant. Not only do chemical reactions
function differently at larger scales, • Reaction heat An API’s particle size has become
but the environmental health & • Reaction initiation temp increasingly important – down
safety (EHS) issues can be enormous. • Adiabatic temp rise to the micron level. Equally as
For instance, how do you safely store • Gas evolution rate important, however, is the Particle
– and eventually dispose of millions Size Distribution (PSD) – essentially,
of gallons of potentially highly-toxic
3. Crystallization how wide the range of particle sizes
chemicals each day? Crystallization often causes problems in a given sample. Common size
if the Critical Process Parameters reduction techniques to meet today’s
Here are five common challenges small, tightly-banded distributions
and their impacts on Critical Quality
in API scale-up Neuland’s scientists include multi-milling, micronization
Attributes (CQA) have not been
frequently confront: or other size reduction (or sometimes
adequately studied. Crystallization
1. Reactions can negatively impact solubility and enlargement) techniques & systems.
dissolution rates. Changes in crystal Where particle size matters
In Unit Processes, reactions may
habit (the external structure) or the (and when doesn’t it?) Particle
negatively impact product selectivity
crystal form (polymorphism – in Engineering studies should be
during scale up if they are not
which the internal arrangement of performed for both drying and
properly understood at the lab scale.
atoms is different) can have wider crystallization using focused beam
This can result in an increase in
impacts on ease of filtration, washing reflectance measurement (FBRM)
impurities – impacting both yield
and drying. and particle vision and measurement
and quality. A thorough working
knowledge of the chemical reactions These changes, in turn, can impact (PVM) probes to understand the
includes the effects of scale- the particle size distribution and impact of process parameters
dependent factors (such as mixing, bulk density, porosity, surface area on target particle size and shape
mass transfer and heat transfer). or polymorph-pseudo polymorph distribution.
Using simulation software (for (solvates/hydrates) to ultimately Determining the proper parameters
example, Dynochem or Visimix), the affect the formulated product’s for API scale-up can be difficult.
effects of mass, heat transfer and drug activity. Process Analytical Using Quality by Design (QbD)
mixing changes can be predicted at Techniques (PAT) can be used during and Design of Experiment (DoE)
larger scales. lab development to understand the approaches, API manufacturers and
impact of process parameters on CDMOs can reduce unanticipated
Design of Experiment (DoE) should
physicochemical properties. challenges by developing deep
be followed at lab scale to develop
a robust process, including the 4. Drying process knowledge at lab scale –
time, temperature and mixing scale which aids in transfer to scale-up.
Drying APIs at larger scales – much
criticalities. This exercise will provide
as with crystallization – can be
adequate Design Space for variability,
complicated. It is common to use
and avoid surprises during scale up &
an NIR probe to perform on-line
technology transfer. The lab process
moisture checks. Residual solvents
validation & qualification to be done
are a frequently-encountered
in the manufacturing plant should
problem during drying, and they are
mimic the conditions at the lab scale.
mainly the result of the crystallization
For example, switching to cylindrical
process. When crystallization
reactors during manufacturing
solvent is trapped in the lattice, it is
may mean slowing the addition of
difficult to remove it to a level that
reagents, reducing the rates at which
meets solvent limits during drying.
heat increases and decreases, and
Crystallization process parameters
more.
should be studied at the lab scale,

15
Contract API Manufacturing: From
Micro-Batches to Multi-Tons
Different Challenges, Different Scales Scale Up Gradually to Identify & Solve When preparing to scale up drug
– The Balancing Act New Challenges at Each Stage production, it’s important to partner
with a CMO well-versed in lifecycle
To some extent, the challenges of Many aspects of early-stage
management. A contract firm that
commercial API manufacturing vary, production, such as handling and
has the extensive expertise needed
based on scale. A large-scale CMO storing raw materials, only become
to work comfortably at all scales
might not necessarily be suited challenging when production
can improve many aspects of the
to working in an early stage of is scaled up. Issues with heat
scale-up process in measurable
development. Applying later-stage generation and dissipation also
ways, making it smoother, faster, and
standards early on, for example, tend to come to the forefront when
easier overall.
could waste time and money – production is dramatically increased.
ultimately derailing the project In addition, larger volumes often The issues that arise when shifting
timeline and budget. mean longer reaction times. This can from one scale to another can be
be problematic for certain processes. unpredictable, and because of this,
At the same time, consideration
planning and forecasting should be
must always be given to later- To detect and resolve these potential
done as early in the R&D process as
stage manufacturing processes & issues, production is scaled
possible. The more experienced the
requirements during earlier phases gradually, going from micro or
transfer team, the more streamlined
of the project to maximize efficiency milligram scale to gram scale, then
and simplified technology transfer
and minimize cost & time-to-market. pilot scale to full production.
can be.
It requires striking a balance –
Lifecycle Management Key to
maximizing early-stage success Several techniques are currently
Successful Process Scale-Up
while minimizing future challenges. used to make production more
efficient. Scouting alternate synthetic and other API manufacturers are • Workplaces become safer overall,
routes to a molecule has become increasingly turning to is green housing lower quantities of toxic
standard, and helps control costs chemistry. solvents and hazardous waste
by accelerating production. This can products.
In green chemistry:
also reduce time spent on regulatory • Processes require less heat,
compliance, as there are fewer steps • Processes are designed to pressure and protective
to document, and less analysis to be maximize the amount of raw equipment.
made throughout the process. materials in usable products
The reduction in hazardous materials
• Operations are designed to use
Consider Your Infrastructure: Analyze and waste also helps decrease the
less energy and to maximize
Processes, Steps, Chemicals & time spent tracking, managing, and
energy efficiency overall
Reagents reporting on safety.
• Environmentally safe or benign
At Neuland, we often manage early products are used whenever While always beneficial for
stage projects in which compounds possible Environmental Health and Safety
are produced in small trial batches. • Waste is avoided as much as (EHS) reasons, green chemistry can
With subsequent scale-up steps possible significantly reduce waste and cost.
on the horizon, however, we In addition, it can also be the key
By following these principles, the
always advise our earlier stage element that enables a drug to be
processes used for larger scale API
clients to consider the discharge, manufactured commercially.
manufacturing can become more
infrastructure and cost of the
efficient. Learn more about Green Chemistry
processes used.
• Reactions use fewer steps, at Neuland.
We systematically analyze how meaning less resources are
each process, step, chemical, or consumed to yield more product.
reagent can potentially affect the • Operations are simpler, and
product when it begins manufacture conducted at ambient temperature
on a larger scale. One solution we and pressure.

17
API Micronization Techniques for Use in
Injectables and Medical Devices
Demand is steadily growing for dissolve more quickly, and allowing compaction using a ball or bead
pharmaceutical materials that effective doses to be injected mill, or other non-conventional
contain micronized active drug in smaller amounts. Poorly- techniques. Each of these techniques
substances (APIs) for inhalation soluble APIs can be improved by has advantages and disadvantages.
and injectable delivery. These active micronization, as it increases surface
For example, bead mills are suitable
pharmaceutical ingredients (APIs) area and speeds up dissolution. For
for heat sensitive materials, but can
are micronized for a number of other dosage forms, micronization
also create some problems such as:
reasons, generally performance- can be used to improve homogeneity
related. in addition to increasing solubility. • Low productivity
• Reliance on large equipment and
For injectable drugs and inhalants, Micronization Defined typically uses an external medium
particle size distribution typically
A particle size of less than 10 for grinding
should be in the range of 2-20
microns can be achieved in a • Potential particulate contamination
microns, with a steep distribution
number of ways, including controlled • Additional processing
curve and minimum amount of fine
precipitation or using an external requirements.
and oversized particles. Depending
source of energy. A conventional
on the complexity of the formulation, Non-Conventional
method of reducing particle size is
device or inhalation delivery system, Micronization
micronization, in which particles
the particle size can vary. However,
are reduced to a size of less than 10 Something to keep in mind is
the majority of particles must be
microns. conventional micronization may not
between 0.5-8 microns.
be suitable for all drug substances.
Micronization typically refers to
With injectable drugs, the particle In the last few years, several
processes that use fluid energy to
size is reduced to increase the supercritical fluid-based techniques
reduce particle size (e.g., jet milling),
solubility of the drug, making it have been proposed to produce

18
micron- and nano-sized particles. achieving bulk density and flowability
Three groups of processes used – critical traits for medical devices.
to produce fine and monodisperse
To fine-tune our techniques,
powders are:
Neuland has conducted particle
• Rapid Expansion of Supercritical size distribution (PSD) trials using
Solutions (RESS) the Design of Experiment model
• Supercritical Antisolvent (SAS) in line with QbD requirements,
• Particles from Gas-Saturated achieving PSD targets at the outer
Solutions (PGSS) limit of the specifications. These
experiments involved identifying,
These techniques are not without
analyzing and fine-tuning a desired
their limitations. By relying on
set of parameters to provide
CO2, they are held back by its poor
accurate, reproducible particle size
solvent power, high cost and the
distributions.
need to use significant amounts.
Despite the number of mechanistic Selecting the appropriate technique
studies available, confusion remains for API micronization remains
concerning how each variable challenging. It is a complex process,
affects the particle morphology and reliant on an experienced team
properties. well-versed in the advantages
and disadvantages of various drug
Common Challenges of
micronization strategies.
Micronization
API Drug Micronization involves many
challenges, risks and considerations.
Some of the most common are:
• The initial particle size of the API to
be micronized – which plays a role in
achieving the desired particle size.
• Reducing particle size also carries
the risk of altering the morphology
of the drug molecule, resulting in
different polymorphs, amorphous
APIs, or a mixture of crystalline and
amorphous APIs.
• Micronized material is charged
and may lead to segregation,
clumping, and other possible physical
instabilities during long term storage
or in the formulation if it is not well
controlled.
API Micronization – Jet Mills
Preferred for Inhalation
Jet mills (or fluidized jet mills) are
the preferred method of micronizing
an API for use in an inhalation dosage
form. Jet milling micronizes drugs
using compressed air or inert gas.
The desired particle size is achieved
by proper control of parameters.
Particle Size Distribution
(PSD) Trials at Neuland
Given our experience with inhalation
products, Neuland has developed
strong micronization capabilities –
providing customers with different
particle size distributions (3-tier
specifications ranging from 2- 5
microns) with a very narrow particle
size range, something that can be
difficult to accomplish. Another
challenge we’ve overcome is

19
Quality & Regulatory
For more than 30 years, Neuland policies ahead of legislation. We have embraced the rise of new
Labs has maintained regulatory quality approaches such as Design
With complex chemical reactions,
excellence – achieving success of Experiment (DoE) and Quality by
quality monitoring takes on an even
in all of our 13 FDA audits and 31 Design (QbD). Our Process Analytical
greater role as products transition
inspections by global regulatory Lab focuses on robust technology
across scale-up or are shifted from
agencies. development and process transfer to
CROs to CMOs.
manufacturing.
Our philosophy is to exceed quality
Neuland has been at the forefront of
levels defined by our customers, In Leveraging QbD for API Scale Up,
research and method development
while meeting stringent international we share how a QbD approach can
for the detection of genotoxic
standards. We continuously monitor be used to understand and control
impurities (GTIs) in drug APIs.
various regulatory bodies worldwide processes based on sound science
Genotoxic Impurities: Increasing
in order to stay ahead of upcoming and quality risk management, while
Vigilance, But Still Some Uncertainty
regulatory changes. This translates How to Strengthen Your API Supply
highlights evolving guidance
into a clear understanding of the Chain Management and Sourcing
surrounding the detection and
possible future direction regulations examines one of today’s most urgent
quantification of GTIs, and discusses
and standards may take, ensuring topics (and a key focus of Neuland
techniques for avoiding their
that we implement appropriate Labs), supply chain security.
formation.

20
Genotoxic Impurities: Increasing
Vigilance, But Still Some Uncertainty
A genotoxic impurity (GTI) is a
chemical substance that can
directly or indirectly damage DNA or
chromosomes and induce genetic
mutations. Fifteen years ago, there
were no specific guidelines for them.
In 2007, however, general awareness
of the risk and consequences of GTIs
surged when Roche’s drug Viracept®
was accidentally contaminated in
a case that quickly became high-
profile. Residual ethanol left in a
storage tank reacted with acid over
a long period of time, creating high
levels of ethyl methane sulfonate
(EMS) that remained in the product.
The EMS levels in the tablets went
undetected until patients who took
them showed adverse effects.
Since then, vigilance concerning
genotoxic impurities has grown, and
regulatory standards have emerged.
The regulatory standards governing
GTIs, however, don’t take long-
term therapeutic usage (potentially
higher, longer-term thresholds) into
account, and some uncertainty exists
regarding several aspects of the
standards.
As some genotoxic impurities cannot multidisciplinary approach should be product materials and components
be completely eliminated, emphasis used. A highly conservative limit, the • facility and equipment operating
is placed on sufficiently controlling TTC level can only be applied singly conditions
them to comply with the impurity to individual GTIs when the impurities • in-process controls
limits set by regulatory bodies. are not structurally similar. • finished product specifications,
Threshold of Toxicological Concern and the associated methods
With multiple structurally-similar
• frequent monitoring and control
For unusually-potent impurities impurities that are expected to act
or those that produce toxic or by similar genotoxic or carcinogenic In a Pharmaceutical Technology
unexpected pharmacological effects, mechanisms, total daily exposure peer-reviewed article, Neuland
the detection and quantification limit should be evaluated in relation to the scientists shared a stability-
of the analysis should match the level TTC. Appropriate individual limits indicating reverse-phase high-
at which the impurities should be should be applied to the sum of all performance liquid chromatographic
controlled. structurally similar GTIs. (RP-HPLC) method for the
quantitative determination of
The Threshold of Toxicological How to Control GTIs
potential genotoxic impurities
Concern (TTC) is the level at To best detect and control GTIs, we present in pemetrexed disodium
which someone can be exposed recommend using a planned set (form IV). The method developed was
to a genotoxic impurity in most of controls – derived from current specific in determining the impurities
pharmaceuticals with minimal product and process understanding at even lower levels than that of
risk, balanced with the therapeutic – that ensures process performance specifications.
benefits of taking the drug. The TTC and product quality. Of paramount
for intake of a genotoxic impurity is importance: selecting a route that
1.5 micrograms per day. Low and doesn’t use genotoxic-alerting
high limits are case-specific and intermediates or reagents – or a
based on each compound’s toxic combination with the potential to
potential. generate GTIs. Controls include:
To assess the potential for GTIs • parameters and attributes related
to affect drug quality, a proactive, to the drug substance and drug

21
Leveraging QbD for API Scale Up
Using Quality by Design (QbD) and • Uni/multi variants With cause & effect analysis of
a Design of Experiments (DoE) • Use of modeling tools Critical Process Parameters (CPP)
approach, API manufacturers and • Use of prior knowledge on Critical Quality Attributes
CDMOs can reduce unanticipated • Control strategy implementation (CQA), QbD also aids in robust
challenges by developing deep • Proactive process monitoring, technology development & transfer
process knowledge at lab scale including PAT for trending, at manufacturing plants. In order
– which aids in transfer to scale- continuous verification and for QbD to improve in scale-up, an
up. QbD can radically transform continuous proactive improvement. appropriate control strategy needs
the scale-up process, and help to be in place to ensure a focus on
Scale-up is a critical link in this
companies avoid unforeseen critical points.
lifecycle. Without enough data, it
complications.
can also be a ‘what if’ exercise. Engineering API Scale-Up
Increased Regulatory Scrutiny Insufficient process knowledge can
The involvement of engineers in
and the Rise of QbD result in poorly scaled processes.
the laboratory during development
This typically translates into more
Regulatory agencies are emphasizing and optimization of a process is
Out Of Spec (OOS) results, reduced
the need for a more thorough key to trouble-free scale-up and
process reliability, as well as higher
understanding of products and technology transfer. The extent to
production costs and a lower
processes prior to validating. This which engineers need to address
profit margin due to increased
has led to widespread adoption of scale-up of operations depends very
reprocessing.
QbD (Quality by Design) approaches, strongly on the interaction between
which emphasize thorough process With development and transfer to the chemist and the engineer, and the
knowledge to avoid poorly-scaled scale-up, the challenges which arise stage at which engineering became
processes. tend to be in a number of different involved in the process.
categories: safety, environmental,
QbD approaches typically use Drug Safety, Efficacy and
health, quality and economics.
a Process Validation Lifecycle Feasibility
Approach, a holistic approach to QbD Enables Robust This collaboration can mean the
development, which supports and Technology Development & difference between a viable drug and
leverages: Transfer to Manufacturing one that had great potential, but was
• Robust validation
not practical from a manufacturing
standpoint. It is customary to
evaluate drugs on two pillars
common to regulatory environments
– efficacy and safety. In other words,
does the drug perform what it needs
to perform, and does it do it safely?
In the real world of drug discovery,
development and commercialization,
however, there is a third equally
important pillar: feasibility.
A product can be determined to be
safe and efficacious – but if it isn’t
feasible to produce (from either an
economic or a technical at-scale
production standpoint), then it isn’t a
candidate for success.
This is especially true since,
often before a scalable chemistry
process has been fully developed,
chromatography (or, more
specifically, process chromatography)
is used for making materials in early-
stage development.

22
Collaborating Across Scales
When chemists and engineers
work hand-in-hand during process Early Development Late Development
development in R&D, processes tend
DRUG DISCOVERY PRODUCT LAUNCH
to progress through scale-up easier.
There are considerable differences Number of >104 20-30 10-15 3-4 2-3
between producing 10 mg batches compounds (Phase I) (Phase II) (Phase III)
and manufacturing 500 kg batches – per year
and numerous engineering-related
factors need to be taken into account. Synthetic 10 mg - 10 g - 1 kg 10 - 100 kg 100 - >1,000 kg
This chart describes the increasing Scale 10 g 1,000 kg
scales in terms of the synthetic Synthetic
EXPEDIENT à PRACTICAL à EFFICIENT à OPTIMAL
process employed – from expedient, Process
to practical, to efficient and –
ultimately – to optimal.
This is the role played by the
collaboration of engineers and
chemists (and the beauty of QbD,
in general): ensuring the smooth
transition from the expedient to the
optimal while developing a safer • identify and characterize any Across the development phase of
process with optimized yield and impurities which have an impact a project, both chemical engineers
quality. on quality. & chemists will work together to
While chemists tend to focus on • perform generation and understand CPPs & CQAs of the
process optimization, engineers qualification of reference/working process.
focus on the scale- and hardware- standards.
More interactions tend to occur
dependent parameters, taking plant • maintain continuous interaction
once process feasibility has been
conditions under consideration. It with IP for process infringement
confirmed and the generated
is this dual approach that enables with any new process patents.
compounds reach a passing level
a ‘Right at First Time’ technology • perform process and method
of quality. Once feasibility has been
transfer. This can be achieved by validation.
shown, the engineers will evaluate
adopting: Chemical Engineers: the process from a safety, health
• A scientific statistical approach and environment standpoint. They
• generate data on the material
(QbD-DOE, ICH Q8), then generate process safety data to
balance.
• Simulation scale-up techniques create inherently safer processes.
• evaluate energy balances to
(Dynochem / Visimix) for scale understand utility requirements for Achieving successful technology
dependent parameters plant scale. transfer requires a robust
• Lab validation of process in • select equipment for commercial manufacturing process, which
conditions simulating the plant scale for retrofitting or new, per – under QbD – is a collaborative
• The use of Quality Risk process requirement. effort, comprising Research and
Assessment (ICH Q9) to evaluate • perform risk assessments (Quality, Development, Manufacturing
each unit process and operations Safety) of unit operations, powder Technical Operations, Quality and
• Development and manufacturing safety characterization studies, more.
(ICH Q10, 11) of robust/safe HAZOP & & HIRA.
processes The benefits of a QbD approach to
• evaluate particle engineering
scale-up are numerous:
Because chemists and chemical (particle size, bulk density, surface
engineers approach each challenge area & Polymorph). • Better manufacturing efficiencies
from different perspectives, multiple • forecast potential for new • Higher yields
areas of expertise are needed. technology implementation • Superior quality
considering the volume of products, • Enhanced process control
Chemists: safety threats, troubleshooting • Higher design space, resulting in
• leverage expertise in various types activities related to the global regulatory flexibility
of synthetic reactions, based on commercial products and more. • Fewer deviations and a scientific
both literature searches and rational for strong CAPA
hands-on experience. Some of the tasks in these lists
• help select the route of synthesis. involve collaboration between the QbD is an effective framework for
• evaluate the feasibility of the two fields. Chemical Engineers, for bringing together a collaborative and
selected route, optimize example, are involved in process inclusive team comprised of both
and validate the process to meet development quite early and also play chemists & engineers to ensure a
the predefined quality and yield. a role in route selection/finalization. successful API scale-up.

23
How to Strengthen Your API Supply
Chain Management and Sourcing

How to Strengthen Your API qualifying API supplier sources, Aim High, with First Time
Supply Chain Management consider these points: Right Approach
and Sourcing • Synthesis route With the goal of continuous
A pharmaceutical manufacturer is • Regulatory standing improvement, employ a First Time
only as good as its supply chain. • Scalability Right (FTR) approach. FTR thinking
Supply chain management and • Cost-efficient processes focuses on suppliers and helping
strategic sourcing are the key drivers At each stage of the process, look for them to avoid potential rejections
which keep operations running sources who meet your expectations – internally though, it aims to focus
smoothly. Understandably, supply for the supply of drug intermediates and solve whatever gaps exist
chain security is one of pharma and key starting materials (KSM) for in specifications, methods and
manufacturing’s most pressing new products. analyses.
issues.
Line Up Multiple Sources By striking a balance between
Set the Right Inventory supplier focus and internal
Targets Once you’ve determined how to seek accountability, this approach can
out and qualify your sources, you can extend to everything from alignment
When planning for the coming use that same process to secure as of test methods including HPLC/GC
quarter, you need to ensure many sources as possible for each columns and release procedures to
capacities are fully met. Adequate KSM. pre-shipment clearance of CoAs. It
inventory of required items
Sources can suddenly be disqualified can also be used to verify the recent
ensures continuous production
or lost without warning due to manufacture of production batches
thereby maximizing manufacturing
unforeseen issues—anything from with select suppliers of your KSM and
capacities.
pollution and business viability to intermediates, confirming the quality
An OTIF – or ‘On Time in Full’ – regulatory issues or other problems of incoming materials is in line with
calculation with a target of 95% which might prompt abrupt closures expectations.
is measured against pre-agreed or supply chain constraints. While
benchmark lead-times. Timeliness
Ensure Regulatory
being caught in a single-source Compliance of the Supplier
of deliveries is measured against situation may be unavoidable (due to
agreed timelines. To achieve a high
Base
specific expertise or infrastructure),
OTIF score means balancing the new alternate suppliers should be A compliance program designed for
cost of non-delivery with the cost of continuously sought and added to KSM and intermediate suppliers can
carrying higher inventory. your source base. Increasing the keep your supplier base audit-ready.
Such a program should emphasize
Choose Pharma Supplier number of trusted sources you can
traceability, and clearly demonstrate
Sources with Care access ensures your company’s
reputation (and reliability) is that all processes are performed
It is critical to identify reliable safeguarded. in compliance with relevant
sources that meet all aspects of regulatory standards and the quality
your company’s requirements. When agreements made with suppliers.

24
Boost Performance
To improve supplier performance
on commercial and technical
parameters, you can measure and
score:
• Supply competitiveness
• Supply quality
• Documentation support
• Compliance
• Speed of supplier response
All of these can be measured on
an objective pre-determined scale,
plotting supply base performance
on two different axes of a Supplier
Performance Dashboard. From there,
Supplier Improvement Actions can be
devised with the goal of moving the
suppliers from three different sub-
optimal quadrants (Q1, Q2 and Q3)
to a more desirable fourth quadrant
(Q4) [see image, at right].
Check Competitiveness
Buying competitiveness can
be measured using relevant
benchmarks. Via a transparent RFQ
process, you can invite multiple
quotes, and then fine-tune the
results based on Commodity Price
Trends.
Deliver On Time VENDOR PERFORMANCE DASHBOARD
A manual tracking program can be
used to track outbound cargo from
origin to destination, per delivery
terms. The Outbound Delivery OTIF,
which is an effectiveness metric, (at
Neuland, for example, the target is
Q3 Q4
Improve on Price,
>=95%) should reflect successful
delivery lead-times vis-à-vis pre-
Payment terms Target Quadrant
determined target lead-times taking & Delivery
into account mode of transport and
destination. Good performance in
OTIF is a key indicator of an end-
to-end supply chain that’s working
effectively.

Q1
By effectively pursuing and gaining
visibility of demand to plan inventory,
choosing and guiding sources that
Q2
Quality

meet your expectations for quality, Improve on Quality,


and keeping everything on track
Service & Compliance
with an OTIF that makes sense for
your business, you can manage
your supply chain and sources with
greater efficiency – improving your
bottom line and benefiting your
customers. Commercial
25
Neuland Laboratories Limited
Sanali Info Park, ‘A’ Block, Ground Floor,
8-2-120/113, Road No. 2, Banjara Hills,
Hyderabad, 500034, Telangana, India
T: +91 40 3021 1600
F: +91 40 3021 1602
E: neuland@neulandlabs.com

You might also like