SlideShare a Scribd company logo
1 of 59
Pharmaceutical
Process Chemistry
SHIKHA D. POPALI
HARSHPAL SINGH WAHI
GURUNANAK COLLEGE OF PHARMACY
INTRODUCTION
2
“Drug” –a compound that interacts with a biological
molecule, triggering a physiological effect.
Drugs (active pharmaceutical ingredients) can be classified
into 4 groups depending on their origin;
1) Natural products
2) Fermentation products
3) Semi-synthetics –substances produced by partial
synthesis
4) Completely synthetic products
Background
 Average cost of developing a drug was $500 million in 1999 and
takes 12-25 years usually.
 The chances of a candidate drug that is identified becoming
approved is approximately 1 in 10,000. (For every 10,000 trial
compounds, 20 reach trials on animals, 10 reach clinical trials on
humans and 1 gets FDA approval)
 Discovery stage (research labs) involves preparation of potential drug
compound on 10 mg to 10 g scale. Toxicology tests are carried out on
this material. If these are successful, greater quantities of the
compound will be required for clinical trials.
 At this stage, development work begins.
-Immediate goal – to produce clinical trial material.
-Longer term goal– to develop a commercially viable scaled-up
validated process.
3
Terminology
 Development–covers all work between research and
production (e.g. analytical, chemical, formulation) and
continues when production begins.
 Scale-up– process of going from laboratory preparation to
whatever scale of manufacture is required to satisfy the
market demand (usually 1,000 to 50,000 L range)
 Technology Transfer
-Transfer between manufacturing sites
-Transfer within a manufacturing site
(Partnerships for collaborative R & D)
4
Process chemistry
5
Process chemistry involves development of practical,
safe and cost effective processes for the synthesis of
compounds selected to progress from
research/discovery to a larger scale.
Within the pharmaceutical industry synthetic routes developed within
medicinal chemistry often have to be completely redesigned by process
chemists, and this is in part due to their differing requirements.
The differences between medicinal and process chemistry and the multitude
of factors that need to be taken into consideration when scaling up a
chemical process.
Medicinal
chemistry Vs
 For medicinal chemists diversity
and flexibility of a route is
important.
 They synthesise a wide range of
analogues of a lead compound
on a small scale (20 mg) for
testing in order to increase
activity, reduce side effects and
to produce an API that may be
easily and efficiently
administered to the patient.
 For this the compound must
have the right properties in
terms of activity, toxicity,
solubility, pharmacokinetics and
pharmacodynamics.
6
 In contrast, process chemistry
involves development of
practical, safe and cost effective
processes for the synthesis of
compounds on a larger scale
(kg to several tonnes) that have
been selected to progress from
medicinal chemistry.
 They generally therefore work
on a single target molecule and
define the best route to that
target.
Process chemistry
Process chemistry
7
The key stages in process chemistry for the development of an active
pharmaceutical ingredient
Process chemistry
8
The key stages in process chemistry for the development of an active
pharmaceutical ingredient
Process chemistry
9
The key stages in process chemistry for the development of an active
pharmaceutical ingredient
DOE in Process Parameters
10
With the most recent ICH and
FDA guidances endorsing a new
paradigm of process validation
based more on process
understanding and control of
parameters and less on product
testing.
After using process knowledgeto relate the attributes to each
process unit operation, the inputs and outputs of each unit
operation are defined to determine process parameters and in-
process controls.
DOE in Process Parameters
11
ICH Q8(R2)provides thefollowingdefinitionsusing thetermcritical:
• Critical process parameter (CPP).A processparameterwhosevariabilityhasanimpactona criticalquality
attributeand,therefore,shouldbemonitoredorcontrolledtoensurethe processproducesthe desired quality.
• Critical qualityattribute (CQA).A physical,chemical, biological,or microbiological propertyor
characteristicthatshouldbewithin anappropriatelimit, range,or distributiontoensurethedesired product
quality.
The definition for CPP states that a parameter is considered critical when its
variability has an impact on a CQA.
The amount of impact is not defined, which leads to the question, does even
a small impact to a CQA mean that the parameter is critical?
DOE in Process Parameters
12
It is not difficult to imagine the example of an extreme shift of a
process parameter having a minor impact on a CQA, whether
measurable or not.
Extreme temperatures can destroy many pharmaceutical
products; however, if a process inherently cannot produce such
temperatures, is temperature still considered to be critical and,
therefore, required to be monitored and controlled?
DOE in Process Parameters
13
When these definitions are strictly interpreted, then one of two
extremes would be:
• Every quality attribute is critical (they all ensure product
quality); every parameter is critical (product cannot be made
without controlling them)
• No parameter is critical because if they are controlled, all
quality attributes will pass specifications.
Reality lies somewhere between these extremes.
Logic and common sense dictate that additional criteria must be
necessary to aid in determining criticality.
There is great value in understanding not only if a
parameter/attribute is critical (i.e., has an impact), but also how
much impact the parameter/attribute has.
Process chemistry
14
The key stages in process chemistry for the development of an active
pharmaceutical ingredient
Process chemistry
15
The key stages in process chemistry for the development of an active
pharmaceutical ingredient
Process chemistry
16
The key stages in process chemistry for the development of an active
pharmaceutical ingredient
Term synthesis means in Greek
“put together”.
Synthetic chemistry is the “art” of building-up
complex molecular structures of compounds
putting together smaller, easily accessible
compounds.
17Synthetic Strategy
Synthetic Strategy 18
The construction of a complex organic structure, defined
target molecule, requires first of all the identification of the
smaller fragments that can be used to build-up the final target.
In the case of oligomers such as peptides,
oligosaccharides or oligonucleotides, the choice is
obvious: the constituent monomers are the building
blocks, and the synthesis requires the junction of those
monomers by condensation.
Two functional groups, one for each monomer, are involved
in the reaction
What is Development?
 Broad range- between research and production- and
overlaps with both
 In API (Active Pharmaceutical Ingredient) manufacture,
begins when active substance is identified and activity
demonstrated and more active substance is required.
 Once a process that works on plant scale is produced and
is validated, development work still continues to improve
cost, efficiency, quality and environmental impact and to
deal with changing circumstances (vendors, equipment,
new regulations
(FDA21 CFR part11, Risk-Based Approach etc))
19
Development Stages
1) Chemical Development
 Begins when activity of potential API is demonstrated
and more active substance is required (for clinical trials).
 Process research;
–New synthetic routes (literature survey)
–Some initial optimisation
–Yield improvements
–Possibly scale-up to large lab equipment/ kilo lab (up to
about 20 L)
20
Development Stages cont’d
2) Process Development
-Optimization (change conditions and parameters)
-Minor change of route / intermediate
-Cheaper / more efficient reagents (new to market)
-Environmentally-friendly reagents and effluent
considerations
-Yield / concentration improvement
-Statistical methods
(e.g. Experimental Design; want maximum amount of unbiased
information about factors affecting a process from as few
observations as possible. Caution -“Facts are stubborn things.
Statistics are more pliable”)
21
Development Stages cont’d
 Process Development cont’d
-SCALE-UP (Kilo Lab, Pilot plant trials)
-Transfer to Manufacturing
Parallel synthesis reactor block
-for optimization of condition
22
Development Stages cont’d
3) Process Support
 Further optimisation (continuous improvement)
 Fine-tuning of yield and throughput
 Cost reduction
 Troubleshooting (reworks, reprocessing,
deviations)
 New vendor approval (use tests)
 Waste minimisation (recycling)
23
Chemical Development
Selection of Synthetic Route
 GET THE BEST ROUTE FROM THE BEGINNING -Difficult to
change later
 Route should be short, efficient, robust and give a high yield
prior to scale-up
 Discovery route often not the best. Expedient not optimal. (raw
materials may not be available in bulk, process may not be
efficient and may be safety hazards on large scale)
 Compare like with like during selection (Same level of
optimisation and same scale. Are products of same purity
obtained? What is the cost comparison?)
 Main constraint is TIME
24
Chemical Development
Selection of Synthetic Route cont’d
 Shortest route usually best (less effluent, shorter lead
time, short plant occupation, less analytical work)
 A convergent synthesis will be cheaper than a
divergent (linear) one with the same number of steps
 Literature not always correct and perseverance with a
reaction is required
 Better to try routes with high chance of success but
those which offer significant benefits should be
attempted even if little precedent
25
Chemical Development
In Convergent Synthesis, sections of the target molecule are prepared and
joined together to form the target molecule. A better yield than a linear
synthesis with the same number of steps will be obtained. The advantage is
significant if the subroutes converge close to the end of the synthesis.
(page 489, “Medicinal Chemistry, An Introduction”, G. Thomas).
26
Process Development
Optimisation of Synthetic Route –Once Selected
 Work up very important
 Can steps easily be combined (telescoping)?
 Need excellent process control and understanding
of process limits (“stress” the reaction –higher
temperature, longer reaction time, less efficient
mixing). “Critical” steps / parameters (Quality)
 Good analytical methods (standards)
 Isolate and characterise by-products
 A very innocuous process change can often have a
significant influence
27
Costing of Processes
 Must be a standard costing procedure
 Should be dated and updated
 Assumptions should be stated
 May be several factors leading to variation –scale,
plant configuration etc.
 Compare like with like and be conservative
 Carried out by independent person
 Want to be able to see which factors increase cost
significantly and why
28
Scale up process
Pharmaceutical Process Scale-Up deals with a
subject both fascinating and vitally important for
the pharmaceutical industry—the procedures of
transferring the results of R&D obtained on
laboratory scale to the pilot plant and finally to
production scale.
29
Why Scale-up?
Scale up is basically needed for:
 Market growth
 Introduction of new processes
 Reduction in making expensive errors in design
and operation
 Concentrate on addressing areas of doubts and
uncertainty
 Economic feasibility
30
Scale-up
Product and process development for scaling up is typically move in small
steps.
 For instance, the development is initially from lab scale to
bench scale then move to pilot scale and finally to
commercialization scale.
 By performing scale up step by step, the risk with large
investments could be lessen.
31
Several
Steps
Application of the SELECT
criteria to scale up
 Safety
 Environmental
 Legal
 Economic
 Control
 Throughput
32
Application of the SELECT
criteria
Safety
Safety is the number one criteria for research
and development. It describes safety risks
including thermal and reactive hazards, and
toxicity, which become increasingly important
as processes are scaled up.
33
Application of the SELECT
criteriaEnvironmental
The pharmaceutical industry follows the
environmental ideals of preventing, minimising and
rendering harmless any reagents used and wastes
generated from processes.
Different metrics are utilised within the industry to
track and minimise environmental impact.
Steps taken to improve environmental credentials for
a process are considered
34
Application of the SELECT
criteriaLegal
It describes legal considerations including
regulation of substances by governments,
environmental legislation and patent law.
35
Application of the SELECT
criteria
Economic
It discusses economic considerations.
36
Application of the SELECT
criteriaControl
Quality control is essential for commercial production
of an API (active pharmaceutical ingredient).
Selectivity, stability, purification, registration and
validation of processes, product specifications and
genotoxic impurities all need to be considered to
ensure quality is suitable for market.
37
Application of the SELECT
criteriaThroughput
Making sure that we can produce enough within a given time, where
chemical yields plays an important role, trying to work as concentrated as
possible; if you have one very diluted step in the middle of synthesis this
can really be a bottleneck.
If you have to perform that several steps before the API this might be real
bottleneck and not allow you to make a sufficient volume.
What's quite often used is a telescoping of reactions, meaning you work in
the same solvent for a number of consecutive steps which is really
improving throughput because
-you don't need to isolate intermediates,
-don't need to filter or centrifugate them,
-you don't need to dry them as
-you don't need to bring them again or dissolve them again for the next
step
So, that's one way to really improve throughput..
38
Summary: Application of the SELECT criteria
39
Criteria Sub-criteria Examples of Potential Issues
Safety
Process safety Explosions
Exposure to harmful substances Carcinogens, sensitisers
Environmental
Wasted resources Quantity & variety of solvents
Substances harmful to the
environment
Aquatic toxins, ozone depleters
Legal
Infringement of Intellectual Property
Competitor patenting key
intermediate
Regulation of reagents and
intermediates
Economic
Meeting Cost of Good (profit
margin)
Long synthesis or expensive starting
materials
Investment costs (equipment)
Control
Control of quality Meeting specifications/ GMP
Control of physical/chemical
parameters
Throughput
Time scale of manufacture Long synthetic route
Availability of starting materials Rare natural products
OBJECT
IVE
40
Stages of Scale up process
41
Stages of Scale up process
Planning for Scale up
 Decide on process
 Decide on batch size (not too large an increase)
 Order raw materials (allow for 10% lower yield)
 Carry out safety tests (Exotherms, HAZOP –hazard and
operability studies)
 Discuss the process and plant requirements with
production manager/engineer (materials of
construction). Existing multipurpose plant usually.
 Prepare safety data sheets and discuss handling of
hazardous reagents or intermediates
 Ensure analytical procedures, equipment and staff are
available
42
Planning for Scale up cont’d
Write out detailed procedure (assume nothing);
 Cleaning check and preparative work
 Charging and weighing
 Temperature and time limits
 Sampling (in-process checks)
 Transfers
 Work-up and isolation
 Drying and effluent disposal/treatment
Make allowance for delays / problems (rework procedures,
identify suitable “hold points”)
43
Planning for Scale up cont’d
Leave space in procedure to record data and
observations
Highlight safety procedures and steps to take if
spillage occurs
Provide training
44
Planning for Scale up cont’d
Significant Differences Between Lab and Plant
 Heat transfer
 Agitation (frothing)
 Mass transfer (affects kinetics)
 Visibility –of reactions, separations, for cleanliness checks
 Separation (stirring, not shaking)
 Time (slower additon rates and heating/cooling times,
longer work up)
 Hazards (Toxic, Exotherm and Electrostatic)
 Off-gas treatment
 Safe and efficient sampling techniques needed (Process
Analytical Technology -Real time reaction monitoring; FT-IR,
Particle size, etc.)
45
Planning for Scale up cont’d
Process Development Taking Account of Differences Between Lab and
Plant Equipment
 Use safety data already produced to help characterise
reaction
 Study effect of scale-dependent factors (mixing, mass
transfer and heat transfer) at lab scale
 Plan a logical set of experiments in time-frame available
 Mimic plant conditions; slower addition of reagents, slower
rate of heat increase and decrease and mixing, lab system
should be baffled if reactors are (no vortex)
 Obtain mass and heat transfer data for reactor type, agitator
type and solvents to be used.
 Use simulation software to predict effects of mass and heat
transfer and mixing changes on scale-up.
46
Essential steps 47
Laboratory Mock-UPS
(Source: Euzen et al., 1993)
Production Capacity
Table below shows the general production capacity of each scaling
up step in the process industries.
48
Scaling factor Typical production
capacity
Bench/ Laboratory 0.001 – 0.1 kg/h
Pilot Plant 10 – 100 kg/h
Demonstration Plant 100 – 1000 kg/h
Commercial Plant > 1000 kg/h
Steps in process
Development
49
 The collection and evaluation of data, from the
process design stage through commercial
production, which establishes scientific evidence that
a process is capable of consistently delivering quality products.
(FDA)
 Documented evidence which provides a high degree of
assurance that a specific process will
consistently result in a product that meets
predetermined specifications and quality
characteristics. (WHO)
 The documented evidence that the process,
operated within established parameters, can
perform effectively and reproducibly to produce a
medicinal product meeting its predetermined
specifications and quality attributes.
50
Validation of Large scale Process
Validation of Large scale Process
Traditional vs new paradigm
Post approval
changes/chan
ge
controls/risk
analysis
Development-
Basic
Process
validation-
3 batches
Pilot batch
manufacturing
Enhanced-
Development
and
process
qualification
Control
Strategy
ICH
Q8,
QbD
Continuous and
extensive
monitoring of
CQAs and
CPPs for each
production
batch
ICH Q9
and Q10
51
52
Pre-validation
phase
Protocol
Preparation
Information from
product
development
studies
(identification of
critical
attributes)
Information from
primary/ clinical
manufacturing
(scale up
information)
Process risk
assessment
information
(identification of
critical steps)
Validation phase
Protocol
execution
Includes
demonstration of
content uniformity
of the clinical
batch
Process validation phases
Post valdn phase:
Review of process,
deviations, failures,
need for
improvement, scale
up etc…
FDA, January 2011 WHO, Revised Annex 7 of
WHO GMP guide (draft for
comment)
European Medicines Agency
(EMA), February 2014
Continuous process
verification (CPV)
Continuous process verification
(CPV)
Alternative approaches:
-Traditional approach
-Continuous process
verification
-Hybrid approach
Process design and Initial
validation (process
qualification- PPQ) are initial
phases of CPV
Process design and initial
validation (initial process
verification) are initial phases of
CPV
CPV protocol to be supported
by extensive dev. information
and lab or pilot scale data.
Executed on each production
batch
No mention of number of
batches for initial process
performance qualification /
validation (rather must be
justified based on overall
product and process
understanding)
Mentions data on at least three
pilot or production batches
collected as part of process
design
Number of batches specified
for traditional approach
- minimum of three production
batches unless other wise
justified
53
Latest guidelines
Prospective validation Concurrent validation Retrospective validation
Protocol reviewed and
accepted, Product PQD; OR
Protocol executed before
submission or PQ
Protocol reviewed and
accepted, Product PQD
Protocol does not need to be
submitted
Execute and finalize process
validation on the first three
production batches
Execute and finalize process
validation on the first three
production batches
Prepare product quality
review report on already
manufactured production
batches
Commercial batches to be
released only after
satisfactorily conclusion of
process validation on three
batches
Each validation batch can be
validated and released.
Applicable for low demand
products (such as NTDs,
orphan drugs or other
seasonal products)
Applicable for submissions
meeting criteria for
established products as
described in Annex 4, TRS
970
54
Types of process validation and
dossier requirements
 Design Qualification( DQ)
verification process to meet
particular requirement relating to
the quality of Pharmaceutical and
manufacturing process.
55
Process validation- Role of assessment
Design
qualificat
ion
Operatio
nal
qualificati
on
Performa
nce
qualificati
on
Process
validatio
n
GMP
Dossier
 Performance qualification (PQ) (process
qualification)- process of testing to ensure that the
individual and combined systems function to meet
agreed performance criteria on a consistent basis
and to check how the result of testing is recorded.
The purpose is to ensure that the criteria specified
can be achieved on a reliable basis over a period of
time.
 Operational Qualification (OQ) process of
testing to ensure individual and combined
systems function to meet agreed performance
criteria and to check how the result of testing
is recorded. The purpose is to ensure that all
the dynamic attributes comply with the
original design.
 DQ plan covers user requirement,
user specification, Technical
specification and DQ report.
 Process Validation :- Owners are responsible for Validating Their Processes (personnel,
equipment, methods, SOPs) to ensure compliance to cGMP/GLP regulations.
Case study; Process Development for
Labetalol Production
BACKGROUND
 Labetalol –antihypertensive, 30,000 Kg p.a. produced by
Schering-Plough.
Labetalol Manufacturing Process
 Mono-pot reaction in jacketed, glass-lined 10,000 L reactor.
 Addition of liquid reagents and jacket temperature computer
controlled. Solid reagents charged manually via handwhole.
 Phase separation using sight-glass.
 Solid product from second step isolated by centrifugation.
 Product tested to determine if recrystallisation necessary.
 When required purity achieved, product is dried
56
Labetalol Process –Step 1
 Solvent system modified to isopropanol / ethyl acetate containing hydrogen
bromide from methanol / ethyl acetate to reduce formation of third impurity.
Original process used chloroform.
 Concentration was increased threefold increasing throughput and reducing
solvent waste.
57
Main Impurities
Labetalol Process – Step 2
Process Development
 Large excess of dibenzylamine used to ensure reaction driven to completion (cheap reagent and easily
washed out)
 Propylene oxide added as it reacts with HBr side-product as it’s produced. Presence of HBr would neutralise
dibenzylamine and no reaction to give product would occur. Propylene bromohydrin side-product easily
washed out.
 Use gentle reflux to ensure propylene oxide doesn’t escape
58
+
Summary
Stages of development are Chemical Development, Process Development and Process
Support
 Development essential before scale-up and validation –get process right
first
 Time constraints a major factor
 Have process in control (“critical” parameters)
 Consider differences between lab and plant scale. Try to mimic plant
conditions in lab to anticipate effects when scale up.
 Prepare process description carefully for scale-up
 Development always on-going
59

More Related Content

What's hot

Halogenation CL, BR, F, I, FREE RADICALS, ADDITION TO ALKENES AND ALYENES
Halogenation CL, BR, F, I, FREE RADICALS, ADDITION TO ALKENES AND ALYENESHalogenation CL, BR, F, I, FREE RADICALS, ADDITION TO ALKENES AND ALYENES
Halogenation CL, BR, F, I, FREE RADICALS, ADDITION TO ALKENES AND ALYENESShikha Popali
 
Strategies for Heterocycle ring synthesis
Strategies for Heterocycle ring synthesis Strategies for Heterocycle ring synthesis
Strategies for Heterocycle ring synthesis SACHINKUMARVISHWAKAR4
 
INTRODUCTION TO PROCESS CHEMISTRY.pptx
INTRODUCTION TO PROCESS CHEMISTRY.pptxINTRODUCTION TO PROCESS CHEMISTRY.pptx
INTRODUCTION TO PROCESS CHEMISTRY.pptxPurushothamKN1
 
Side reaction in peptide synthesis
Side reaction in peptide synthesisSide reaction in peptide synthesis
Side reaction in peptide synthesissuraj wanjari
 
Heterocyclic compounds
Heterocyclic compoundsHeterocyclic compounds
Heterocyclic compoundspriyaswain27
 
Homogenous Catalysis
Homogenous CatalysisHomogenous Catalysis
Homogenous CatalysisDivya V
 
CONTINUOUS FLOW REACTORS (1).pptx
CONTINUOUS FLOW REACTORS (1).pptxCONTINUOUS FLOW REACTORS (1).pptx
CONTINUOUS FLOW REACTORS (1).pptxupasanasharma66
 
Chemistry of peptide (BPHARM,MPHARM,MSC,BSC)
Chemistry of peptide (BPHARM,MPHARM,MSC,BSC)Chemistry of peptide (BPHARM,MPHARM,MSC,BSC)
Chemistry of peptide (BPHARM,MPHARM,MSC,BSC)Shikha Popali
 
Homogenous catalysis & Biocatalysis
Homogenous catalysis & BiocatalysisHomogenous catalysis & Biocatalysis
Homogenous catalysis & Biocatalysiskavyakaparthi1
 
Oxidation (Unit Process-1) Pharmaceutical Process Chemistry
Oxidation (Unit Process-1) Pharmaceutical Process Chemistry Oxidation (Unit Process-1) Pharmaceutical Process Chemistry
Oxidation (Unit Process-1) Pharmaceutical Process Chemistry AikanGupta
 
UNIT OPERATIONS (unit 2) pharmaceutical process chemistry
UNIT OPERATIONS (unit 2) pharmaceutical process chemistryUNIT OPERATIONS (unit 2) pharmaceutical process chemistry
UNIT OPERATIONS (unit 2) pharmaceutical process chemistrySaketChoudhary13
 
Agents acting on HIV Protease enzyme
Agents acting on HIV Protease enzymeAgents acting on HIV Protease enzyme
Agents acting on HIV Protease enzymeLakshayGupta67
 
MOLECULAR DOCKING AND DRUG RECEPTOR INTERACTION AGENT ACTING.pptx
MOLECULAR DOCKING AND DRUG RECEPTOR INTERACTION AGENT ACTING.pptxMOLECULAR DOCKING AND DRUG RECEPTOR INTERACTION AGENT ACTING.pptx
MOLECULAR DOCKING AND DRUG RECEPTOR INTERACTION AGENT ACTING.pptxMO.SHAHANAWAZ
 
STEREOCHEMISTRY AND DRUG ACTION.pptx
STEREOCHEMISTRY AND DRUG ACTION.pptxSTEREOCHEMISTRY AND DRUG ACTION.pptx
STEREOCHEMISTRY AND DRUG ACTION.pptxAchalYawalkar
 
Continuous flow reaction/ Chemistry
Continuous flow reaction/ ChemistryContinuous flow reaction/ Chemistry
Continuous flow reaction/ ChemistryGagangowda58
 

What's hot (20)

Halogenation CL, BR, F, I, FREE RADICALS, ADDITION TO ALKENES AND ALYENES
Halogenation CL, BR, F, I, FREE RADICALS, ADDITION TO ALKENES AND ALYENESHalogenation CL, BR, F, I, FREE RADICALS, ADDITION TO ALKENES AND ALYENES
Halogenation CL, BR, F, I, FREE RADICALS, ADDITION TO ALKENES AND ALYENES
 
Nitration (2)
Nitration (2)Nitration (2)
Nitration (2)
 
Reduction Reaction
Reduction Reaction  Reduction Reaction
Reduction Reaction
 
Strategies for Heterocycle ring synthesis
Strategies for Heterocycle ring synthesis Strategies for Heterocycle ring synthesis
Strategies for Heterocycle ring synthesis
 
INTRODUCTION TO PROCESS CHEMISTRY.pptx
INTRODUCTION TO PROCESS CHEMISTRY.pptxINTRODUCTION TO PROCESS CHEMISTRY.pptx
INTRODUCTION TO PROCESS CHEMISTRY.pptx
 
Side reaction in peptide synthesis
Side reaction in peptide synthesisSide reaction in peptide synthesis
Side reaction in peptide synthesis
 
synthon approach
 synthon approach  synthon approach
synthon approach
 
Heterocyclic compounds
Heterocyclic compoundsHeterocyclic compounds
Heterocyclic compounds
 
Homogenous Catalysis
Homogenous CatalysisHomogenous Catalysis
Homogenous Catalysis
 
CONTINUOUS FLOW REACTORS (1).pptx
CONTINUOUS FLOW REACTORS (1).pptxCONTINUOUS FLOW REACTORS (1).pptx
CONTINUOUS FLOW REACTORS (1).pptx
 
Impurities in API , types and their sources including genotoxic impurities
Impurities in API , types and their sources including genotoxic impuritiesImpurities in API , types and their sources including genotoxic impurities
Impurities in API , types and their sources including genotoxic impurities
 
Chemistry of peptide (BPHARM,MPHARM,MSC,BSC)
Chemistry of peptide (BPHARM,MPHARM,MSC,BSC)Chemistry of peptide (BPHARM,MPHARM,MSC,BSC)
Chemistry of peptide (BPHARM,MPHARM,MSC,BSC)
 
Homogenous catalysis & Biocatalysis
Homogenous catalysis & BiocatalysisHomogenous catalysis & Biocatalysis
Homogenous catalysis & Biocatalysis
 
Oxidation (Unit Process-1) Pharmaceutical Process Chemistry
Oxidation (Unit Process-1) Pharmaceutical Process Chemistry Oxidation (Unit Process-1) Pharmaceutical Process Chemistry
Oxidation (Unit Process-1) Pharmaceutical Process Chemistry
 
UNIT OPERATIONS (unit 2) pharmaceutical process chemistry
UNIT OPERATIONS (unit 2) pharmaceutical process chemistryUNIT OPERATIONS (unit 2) pharmaceutical process chemistry
UNIT OPERATIONS (unit 2) pharmaceutical process chemistry
 
Agents acting on HIV Protease enzyme
Agents acting on HIV Protease enzymeAgents acting on HIV Protease enzyme
Agents acting on HIV Protease enzyme
 
MOLECULAR DOCKING AND DRUG RECEPTOR INTERACTION AGENT ACTING.pptx
MOLECULAR DOCKING AND DRUG RECEPTOR INTERACTION AGENT ACTING.pptxMOLECULAR DOCKING AND DRUG RECEPTOR INTERACTION AGENT ACTING.pptx
MOLECULAR DOCKING AND DRUG RECEPTOR INTERACTION AGENT ACTING.pptx
 
STEREOCHEMISTRY AND DRUG ACTION.pptx
STEREOCHEMISTRY AND DRUG ACTION.pptxSTEREOCHEMISTRY AND DRUG ACTION.pptx
STEREOCHEMISTRY AND DRUG ACTION.pptx
 
Continuous flow reaction/ Chemistry
Continuous flow reaction/ ChemistryContinuous flow reaction/ Chemistry
Continuous flow reaction/ Chemistry
 
Reduction
ReductionReduction
Reduction
 

Similar to Process chemistry AS PER PCI SYLLABUS FOR M.PHARM

The Path Ahead_ Continuous Processing in API Pharmaceutical Manufacturing
The Path Ahead_ Continuous Processing in API Pharmaceutical ManufacturingThe Path Ahead_ Continuous Processing in API Pharmaceutical Manufacturing
The Path Ahead_ Continuous Processing in API Pharmaceutical ManufacturingMutyala Naidu G
 
ICH Q8 " PHARMACEUTICAL DEVELOPMENT"
ICH Q8 " PHARMACEUTICAL DEVELOPMENT"ICH Q8 " PHARMACEUTICAL DEVELOPMENT"
ICH Q8 " PHARMACEUTICAL DEVELOPMENT"Talha Mahmood
 
MPH14-Quality by Design Salunkhe..pptx
MPH14-Quality by Design  Salunkhe..pptxMPH14-Quality by Design  Salunkhe..pptx
MPH14-Quality by Design Salunkhe..pptxVinayshriSalunkhe
 
ICH Q8 GUIDELINES OF QUALITY BY DESIGN(PRODUCT DEVELOPEMENT)
ICH Q8 GUIDELINES OF QUALITY BY DESIGN(PRODUCT DEVELOPEMENT)ICH Q8 GUIDELINES OF QUALITY BY DESIGN(PRODUCT DEVELOPEMENT)
ICH Q8 GUIDELINES OF QUALITY BY DESIGN(PRODUCT DEVELOPEMENT)ROHIT
 
Microchemistry
Microchemistry Microchemistry
Microchemistry Trisha Das
 
Quality by design in pharmaceutical development
Quality by design in pharmaceutical developmentQuality by design in pharmaceutical development
Quality by design in pharmaceutical developmentManish Rajput
 
Scale Up Methodology for the Fine Chemical Industry - The Influence of the Mi...
Scale Up Methodology for the Fine Chemical Industry - The Influence of the Mi...Scale Up Methodology for the Fine Chemical Industry - The Influence of the Mi...
Scale Up Methodology for the Fine Chemical Industry - The Influence of the Mi...Aldo Shusterman
 
Scale Up Methodology for the Fine Chemical Industry - The Influence of the Mi...
Scale Up Methodology for the Fine Chemical Industry - The Influence of the Mi...Scale Up Methodology for the Fine Chemical Industry - The Influence of the Mi...
Scale Up Methodology for the Fine Chemical Industry - The Influence of the Mi...Aldo Shusterman
 
Anthony crasto scaleup techniques & pilot plant
Anthony crasto scaleup techniques & pilot plantAnthony crasto scaleup techniques & pilot plant
Anthony crasto scaleup techniques & pilot plantAnthony Melvin Crasto Ph.D
 
Green chemistry in pharmaceuticals
Green chemistry in pharmaceuticalsGreen chemistry in pharmaceuticals
Green chemistry in pharmaceuticalsKapish Kapoor
 
CRO Company for Mode of Action & Efficacy Test for Drugs | InvivoBiosystems
CRO Company for Mode of Action & Efficacy Test for Drugs | InvivoBiosystemsCRO Company for Mode of Action & Efficacy Test for Drugs | InvivoBiosystems
CRO Company for Mode of Action & Efficacy Test for Drugs | InvivoBiosystemsWenlan Hu
 
Best compound characterization protocol
Best compound characterization protocolBest compound characterization protocol
Best compound characterization protocolWenlan Hu
 
Quality-By-Design In Pharmaceutical Development
Quality-By-Design In Pharmaceutical DevelopmentQuality-By-Design In Pharmaceutical Development
Quality-By-Design In Pharmaceutical DevelopmentShubhangiParbhane1
 
Development and Scale Up Of a Chemical Process in Pharmaceutical Industry: A ...
Development and Scale Up Of a Chemical Process in Pharmaceutical Industry: A ...Development and Scale Up Of a Chemical Process in Pharmaceutical Industry: A ...
Development and Scale Up Of a Chemical Process in Pharmaceutical Industry: A ...IJERA Editor
 
Targeting Factors of Ecotax Based on Life Cycle Assessment for Select Criteri...
Targeting Factors of Ecotax Based on Life Cycle Assessment for Select Criteri...Targeting Factors of Ecotax Based on Life Cycle Assessment for Select Criteri...
Targeting Factors of Ecotax Based on Life Cycle Assessment for Select Criteri...AI Publications
 

Similar to Process chemistry AS PER PCI SYLLABUS FOR M.PHARM (20)

Optimization 1
Optimization 1Optimization 1
Optimization 1
 
Process research overview
Process research  overviewProcess research  overview
Process research overview
 
The Path Ahead_ Continuous Processing in API Pharmaceutical Manufacturing
The Path Ahead_ Continuous Processing in API Pharmaceutical ManufacturingThe Path Ahead_ Continuous Processing in API Pharmaceutical Manufacturing
The Path Ahead_ Continuous Processing in API Pharmaceutical Manufacturing
 
ICH Q8
ICH Q8ICH Q8
ICH Q8
 
ICH Q8 " PHARMACEUTICAL DEVELOPMENT"
ICH Q8 " PHARMACEUTICAL DEVELOPMENT"ICH Q8 " PHARMACEUTICAL DEVELOPMENT"
ICH Q8 " PHARMACEUTICAL DEVELOPMENT"
 
Ich q8
Ich q8Ich q8
Ich q8
 
MPH14-Quality by Design Salunkhe..pptx
MPH14-Quality by Design  Salunkhe..pptxMPH14-Quality by Design  Salunkhe..pptx
MPH14-Quality by Design Salunkhe..pptx
 
ICH Q8 GUIDELINES OF QUALITY BY DESIGN(PRODUCT DEVELOPEMENT)
ICH Q8 GUIDELINES OF QUALITY BY DESIGN(PRODUCT DEVELOPEMENT)ICH Q8 GUIDELINES OF QUALITY BY DESIGN(PRODUCT DEVELOPEMENT)
ICH Q8 GUIDELINES OF QUALITY BY DESIGN(PRODUCT DEVELOPEMENT)
 
Microchemistry
Microchemistry Microchemistry
Microchemistry
 
Quality by design in pharmaceutical development
Quality by design in pharmaceutical developmentQuality by design in pharmaceutical development
Quality by design in pharmaceutical development
 
Scale Up Methodology for the Fine Chemical Industry - The Influence of the Mi...
Scale Up Methodology for the Fine Chemical Industry - The Influence of the Mi...Scale Up Methodology for the Fine Chemical Industry - The Influence of the Mi...
Scale Up Methodology for the Fine Chemical Industry - The Influence of the Mi...
 
Scale Up Methodology for the Fine Chemical Industry - The Influence of the Mi...
Scale Up Methodology for the Fine Chemical Industry - The Influence of the Mi...Scale Up Methodology for the Fine Chemical Industry - The Influence of the Mi...
Scale Up Methodology for the Fine Chemical Industry - The Influence of the Mi...
 
Anthony crasto scaleup techniques & pilot plant
Anthony crasto scaleup techniques & pilot plantAnthony crasto scaleup techniques & pilot plant
Anthony crasto scaleup techniques & pilot plant
 
Green chemistry in pharmaceuticals
Green chemistry in pharmaceuticalsGreen chemistry in pharmaceuticals
Green chemistry in pharmaceuticals
 
CRO Company for Mode of Action & Efficacy Test for Drugs | InvivoBiosystems
CRO Company for Mode of Action & Efficacy Test for Drugs | InvivoBiosystemsCRO Company for Mode of Action & Efficacy Test for Drugs | InvivoBiosystems
CRO Company for Mode of Action & Efficacy Test for Drugs | InvivoBiosystems
 
Best compound characterization protocol
Best compound characterization protocolBest compound characterization protocol
Best compound characterization protocol
 
Drug discovery hit to lead
Drug discovery hit to leadDrug discovery hit to lead
Drug discovery hit to lead
 
Quality-By-Design In Pharmaceutical Development
Quality-By-Design In Pharmaceutical DevelopmentQuality-By-Design In Pharmaceutical Development
Quality-By-Design In Pharmaceutical Development
 
Development and Scale Up Of a Chemical Process in Pharmaceutical Industry: A ...
Development and Scale Up Of a Chemical Process in Pharmaceutical Industry: A ...Development and Scale Up Of a Chemical Process in Pharmaceutical Industry: A ...
Development and Scale Up Of a Chemical Process in Pharmaceutical Industry: A ...
 
Targeting Factors of Ecotax Based on Life Cycle Assessment for Select Criteri...
Targeting Factors of Ecotax Based on Life Cycle Assessment for Select Criteri...Targeting Factors of Ecotax Based on Life Cycle Assessment for Select Criteri...
Targeting Factors of Ecotax Based on Life Cycle Assessment for Select Criteri...
 

More from Shikha Popali

Steric parameters taft’s steric factor (es)
Steric parameters  taft’s steric factor (es)Steric parameters  taft’s steric factor (es)
Steric parameters taft’s steric factor (es)Shikha Popali
 
Stereochemistry in drug design
Stereochemistry in drug designStereochemistry in drug design
Stereochemistry in drug designShikha Popali
 
Bioanalytical method validation
Bioanalytical method validationBioanalytical method validation
Bioanalytical method validationShikha Popali
 
Antihypertensive drugs
Antihypertensive drugsAntihypertensive drugs
Antihypertensive drugsShikha Popali
 
Herbal ingredient hair and skin care
Herbal ingredient hair and skin careHerbal ingredient hair and skin care
Herbal ingredient hair and skin careShikha Popali
 
problem assso with oral cavity
problem assso with oral cavityproblem assso with oral cavity
problem assso with oral cavityShikha Popali
 
problems associated with skin[ as per Pharmaceutics]
problems associated with skin[ as per Pharmaceutics]problems associated with skin[ as per Pharmaceutics]
problems associated with skin[ as per Pharmaceutics]Shikha Popali
 
cosmetics and cosmeticals [department of pharmaceutics]
cosmetics and cosmeticals [department of pharmaceutics]cosmetics and cosmeticals [department of pharmaceutics]
cosmetics and cosmeticals [department of pharmaceutics]Shikha Popali
 
MUTUAL PRODRUG [PHARMACEUTICALS]
MUTUAL PRODRUG [PHARMACEUTICALS]MUTUAL PRODRUG [PHARMACEUTICALS]
MUTUAL PRODRUG [PHARMACEUTICALS]Shikha Popali
 
TITANIUM CHLORIDE [PHARMACEUTICAL REAGENT]
TITANIUM CHLORIDE [PHARMACEUTICAL REAGENT]TITANIUM CHLORIDE [PHARMACEUTICAL REAGENT]
TITANIUM CHLORIDE [PHARMACEUTICAL REAGENT]Shikha Popali
 
OLIGONUCLEOTIDE THERAPY [ TECHNIQUES, APPLICATIONS]
OLIGONUCLEOTIDE THERAPY [ TECHNIQUES, APPLICATIONS]OLIGONUCLEOTIDE THERAPY [ TECHNIQUES, APPLICATIONS]
OLIGONUCLEOTIDE THERAPY [ TECHNIQUES, APPLICATIONS]Shikha Popali
 
OXIDATION [PHARMACEUTICAL PROCESS CHEMISTRY]
OXIDATION [PHARMACEUTICAL PROCESS CHEMISTRY]OXIDATION [PHARMACEUTICAL PROCESS CHEMISTRY]
OXIDATION [PHARMACEUTICAL PROCESS CHEMISTRY]Shikha Popali
 
Synthetic reagent and applications OF ALUMINIUM ISOPROPOXIDE
Synthetic reagent and applications OF ALUMINIUM ISOPROPOXIDESynthetic reagent and applications OF ALUMINIUM ISOPROPOXIDE
Synthetic reagent and applications OF ALUMINIUM ISOPROPOXIDEShikha Popali
 
PHASE TRANSFER CATALYSIS [PTC]
PHASE TRANSFER CATALYSIS [PTC] PHASE TRANSFER CATALYSIS [PTC]
PHASE TRANSFER CATALYSIS [PTC] Shikha Popali
 
SWERTIA CHIRATA NATURAL PRODUCT OF PHARMACEUTICALS
SWERTIA CHIRATA  NATURAL PRODUCT OF PHARMACEUTICALSSWERTIA CHIRATA  NATURAL PRODUCT OF PHARMACEUTICALS
SWERTIA CHIRATA NATURAL PRODUCT OF PHARMACEUTICALSShikha Popali
 
LIQUID CHROMATOGRAPHY- MASS SPECTROSCOPY[LC-MS]
LIQUID CHROMATOGRAPHY- MASS SPECTROSCOPY[LC-MS]LIQUID CHROMATOGRAPHY- MASS SPECTROSCOPY[LC-MS]
LIQUID CHROMATOGRAPHY- MASS SPECTROSCOPY[LC-MS]Shikha Popali
 
EVAPORATION (LIQUID CONVERSION INTO VAPOUR PRESSURE)
EVAPORATION (LIQUID CONVERSION INTO VAPOUR PRESSURE)EVAPORATION (LIQUID CONVERSION INTO VAPOUR PRESSURE)
EVAPORATION (LIQUID CONVERSION INTO VAPOUR PRESSURE)Shikha Popali
 
Homogeneous catalysis [ MPHARM, MSC, BPHARM, BSC]
Homogeneous catalysis [ MPHARM, MSC, BPHARM, BSC]Homogeneous catalysis [ MPHARM, MSC, BPHARM, BSC]
Homogeneous catalysis [ MPHARM, MSC, BPHARM, BSC]Shikha Popali
 
Dicyclohexylcarbodiimide [DCC]
Dicyclohexylcarbodiimide [DCC]Dicyclohexylcarbodiimide [DCC]
Dicyclohexylcarbodiimide [DCC]Shikha Popali
 
COMPARATIVE EVALUATION OF DIFFERENT PARACETAMOL BRANDS
COMPARATIVE EVALUATION OF DIFFERENT PARACETAMOL BRANDSCOMPARATIVE EVALUATION OF DIFFERENT PARACETAMOL BRANDS
COMPARATIVE EVALUATION OF DIFFERENT PARACETAMOL BRANDSShikha Popali
 

More from Shikha Popali (20)

Steric parameters taft’s steric factor (es)
Steric parameters  taft’s steric factor (es)Steric parameters  taft’s steric factor (es)
Steric parameters taft’s steric factor (es)
 
Stereochemistry in drug design
Stereochemistry in drug designStereochemistry in drug design
Stereochemistry in drug design
 
Bioanalytical method validation
Bioanalytical method validationBioanalytical method validation
Bioanalytical method validation
 
Antihypertensive drugs
Antihypertensive drugsAntihypertensive drugs
Antihypertensive drugs
 
Herbal ingredient hair and skin care
Herbal ingredient hair and skin careHerbal ingredient hair and skin care
Herbal ingredient hair and skin care
 
problem assso with oral cavity
problem assso with oral cavityproblem assso with oral cavity
problem assso with oral cavity
 
problems associated with skin[ as per Pharmaceutics]
problems associated with skin[ as per Pharmaceutics]problems associated with skin[ as per Pharmaceutics]
problems associated with skin[ as per Pharmaceutics]
 
cosmetics and cosmeticals [department of pharmaceutics]
cosmetics and cosmeticals [department of pharmaceutics]cosmetics and cosmeticals [department of pharmaceutics]
cosmetics and cosmeticals [department of pharmaceutics]
 
MUTUAL PRODRUG [PHARMACEUTICALS]
MUTUAL PRODRUG [PHARMACEUTICALS]MUTUAL PRODRUG [PHARMACEUTICALS]
MUTUAL PRODRUG [PHARMACEUTICALS]
 
TITANIUM CHLORIDE [PHARMACEUTICAL REAGENT]
TITANIUM CHLORIDE [PHARMACEUTICAL REAGENT]TITANIUM CHLORIDE [PHARMACEUTICAL REAGENT]
TITANIUM CHLORIDE [PHARMACEUTICAL REAGENT]
 
OLIGONUCLEOTIDE THERAPY [ TECHNIQUES, APPLICATIONS]
OLIGONUCLEOTIDE THERAPY [ TECHNIQUES, APPLICATIONS]OLIGONUCLEOTIDE THERAPY [ TECHNIQUES, APPLICATIONS]
OLIGONUCLEOTIDE THERAPY [ TECHNIQUES, APPLICATIONS]
 
OXIDATION [PHARMACEUTICAL PROCESS CHEMISTRY]
OXIDATION [PHARMACEUTICAL PROCESS CHEMISTRY]OXIDATION [PHARMACEUTICAL PROCESS CHEMISTRY]
OXIDATION [PHARMACEUTICAL PROCESS CHEMISTRY]
 
Synthetic reagent and applications OF ALUMINIUM ISOPROPOXIDE
Synthetic reagent and applications OF ALUMINIUM ISOPROPOXIDESynthetic reagent and applications OF ALUMINIUM ISOPROPOXIDE
Synthetic reagent and applications OF ALUMINIUM ISOPROPOXIDE
 
PHASE TRANSFER CATALYSIS [PTC]
PHASE TRANSFER CATALYSIS [PTC] PHASE TRANSFER CATALYSIS [PTC]
PHASE TRANSFER CATALYSIS [PTC]
 
SWERTIA CHIRATA NATURAL PRODUCT OF PHARMACEUTICALS
SWERTIA CHIRATA  NATURAL PRODUCT OF PHARMACEUTICALSSWERTIA CHIRATA  NATURAL PRODUCT OF PHARMACEUTICALS
SWERTIA CHIRATA NATURAL PRODUCT OF PHARMACEUTICALS
 
LIQUID CHROMATOGRAPHY- MASS SPECTROSCOPY[LC-MS]
LIQUID CHROMATOGRAPHY- MASS SPECTROSCOPY[LC-MS]LIQUID CHROMATOGRAPHY- MASS SPECTROSCOPY[LC-MS]
LIQUID CHROMATOGRAPHY- MASS SPECTROSCOPY[LC-MS]
 
EVAPORATION (LIQUID CONVERSION INTO VAPOUR PRESSURE)
EVAPORATION (LIQUID CONVERSION INTO VAPOUR PRESSURE)EVAPORATION (LIQUID CONVERSION INTO VAPOUR PRESSURE)
EVAPORATION (LIQUID CONVERSION INTO VAPOUR PRESSURE)
 
Homogeneous catalysis [ MPHARM, MSC, BPHARM, BSC]
Homogeneous catalysis [ MPHARM, MSC, BPHARM, BSC]Homogeneous catalysis [ MPHARM, MSC, BPHARM, BSC]
Homogeneous catalysis [ MPHARM, MSC, BPHARM, BSC]
 
Dicyclohexylcarbodiimide [DCC]
Dicyclohexylcarbodiimide [DCC]Dicyclohexylcarbodiimide [DCC]
Dicyclohexylcarbodiimide [DCC]
 
COMPARATIVE EVALUATION OF DIFFERENT PARACETAMOL BRANDS
COMPARATIVE EVALUATION OF DIFFERENT PARACETAMOL BRANDSCOMPARATIVE EVALUATION OF DIFFERENT PARACETAMOL BRANDS
COMPARATIVE EVALUATION OF DIFFERENT PARACETAMOL BRANDS
 

Recently uploaded

Earth Day Presentation wow hello nice great
Earth Day Presentation wow hello nice greatEarth Day Presentation wow hello nice great
Earth Day Presentation wow hello nice greatYousafMalik24
 
Software Engineering Methodologies (overview)
Software Engineering Methodologies (overview)Software Engineering Methodologies (overview)
Software Engineering Methodologies (overview)eniolaolutunde
 
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPTECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPTiammrhaywood
 
Computed Fields and api Depends in the Odoo 17
Computed Fields and api Depends in the Odoo 17Computed Fields and api Depends in the Odoo 17
Computed Fields and api Depends in the Odoo 17Celine George
 
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...Marc Dusseiller Dusjagr
 
Pharmacognosy Flower 3. Compositae 2023.pdf
Pharmacognosy Flower 3. Compositae 2023.pdfPharmacognosy Flower 3. Compositae 2023.pdf
Pharmacognosy Flower 3. Compositae 2023.pdfMahmoud M. Sallam
 
Framing an Appropriate Research Question 6b9b26d93da94caf993c038d9efcdedb.pdf
Framing an Appropriate Research Question 6b9b26d93da94caf993c038d9efcdedb.pdfFraming an Appropriate Research Question 6b9b26d93da94caf993c038d9efcdedb.pdf
Framing an Appropriate Research Question 6b9b26d93da94caf993c038d9efcdedb.pdfUjwalaBharambe
 
EPANDING THE CONTENT OF AN OUTLINE using notes.pptx
EPANDING THE CONTENT OF AN OUTLINE using notes.pptxEPANDING THE CONTENT OF AN OUTLINE using notes.pptx
EPANDING THE CONTENT OF AN OUTLINE using notes.pptxRaymartEstabillo3
 
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️9953056974 Low Rate Call Girls In Saket, Delhi NCR
 
Painted Grey Ware.pptx, PGW Culture of India
Painted Grey Ware.pptx, PGW Culture of IndiaPainted Grey Ware.pptx, PGW Culture of India
Painted Grey Ware.pptx, PGW Culture of IndiaVirag Sontakke
 
Solving Puzzles Benefits Everyone (English).pptx
Solving Puzzles Benefits Everyone (English).pptxSolving Puzzles Benefits Everyone (English).pptx
Solving Puzzles Benefits Everyone (English).pptxOH TEIK BIN
 
How to Configure Email Server in Odoo 17
How to Configure Email Server in Odoo 17How to Configure Email Server in Odoo 17
How to Configure Email Server in Odoo 17Celine George
 
DATA STRUCTURE AND ALGORITHM for beginners
DATA STRUCTURE AND ALGORITHM for beginnersDATA STRUCTURE AND ALGORITHM for beginners
DATA STRUCTURE AND ALGORITHM for beginnersSabitha Banu
 
Presiding Officer Training module 2024 lok sabha elections
Presiding Officer Training module 2024 lok sabha electionsPresiding Officer Training module 2024 lok sabha elections
Presiding Officer Training module 2024 lok sabha electionsanshu789521
 
Introduction to AI in Higher Education_draft.pptx
Introduction to AI in Higher Education_draft.pptxIntroduction to AI in Higher Education_draft.pptx
Introduction to AI in Higher Education_draft.pptxpboyjonauth
 
History Class XII Ch. 3 Kinship, Caste and Class (1).pptx
History Class XII Ch. 3 Kinship, Caste and Class (1).pptxHistory Class XII Ch. 3 Kinship, Caste and Class (1).pptx
History Class XII Ch. 3 Kinship, Caste and Class (1).pptxsocialsciencegdgrohi
 
Biting mechanism of poisonous snakes.pdf
Biting mechanism of poisonous snakes.pdfBiting mechanism of poisonous snakes.pdf
Biting mechanism of poisonous snakes.pdfadityarao40181
 

Recently uploaded (20)

Earth Day Presentation wow hello nice great
Earth Day Presentation wow hello nice greatEarth Day Presentation wow hello nice great
Earth Day Presentation wow hello nice great
 
Software Engineering Methodologies (overview)
Software Engineering Methodologies (overview)Software Engineering Methodologies (overview)
Software Engineering Methodologies (overview)
 
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPTECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
 
Computed Fields and api Depends in the Odoo 17
Computed Fields and api Depends in the Odoo 17Computed Fields and api Depends in the Odoo 17
Computed Fields and api Depends in the Odoo 17
 
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...
 
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
 
Pharmacognosy Flower 3. Compositae 2023.pdf
Pharmacognosy Flower 3. Compositae 2023.pdfPharmacognosy Flower 3. Compositae 2023.pdf
Pharmacognosy Flower 3. Compositae 2023.pdf
 
Framing an Appropriate Research Question 6b9b26d93da94caf993c038d9efcdedb.pdf
Framing an Appropriate Research Question 6b9b26d93da94caf993c038d9efcdedb.pdfFraming an Appropriate Research Question 6b9b26d93da94caf993c038d9efcdedb.pdf
Framing an Appropriate Research Question 6b9b26d93da94caf993c038d9efcdedb.pdf
 
EPANDING THE CONTENT OF AN OUTLINE using notes.pptx
EPANDING THE CONTENT OF AN OUTLINE using notes.pptxEPANDING THE CONTENT OF AN OUTLINE using notes.pptx
EPANDING THE CONTENT OF AN OUTLINE using notes.pptx
 
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️
 
Painted Grey Ware.pptx, PGW Culture of India
Painted Grey Ware.pptx, PGW Culture of IndiaPainted Grey Ware.pptx, PGW Culture of India
Painted Grey Ware.pptx, PGW Culture of India
 
Solving Puzzles Benefits Everyone (English).pptx
Solving Puzzles Benefits Everyone (English).pptxSolving Puzzles Benefits Everyone (English).pptx
Solving Puzzles Benefits Everyone (English).pptx
 
Model Call Girl in Bikash Puri Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Bikash Puri  Delhi reach out to us at 🔝9953056974🔝Model Call Girl in Bikash Puri  Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Bikash Puri Delhi reach out to us at 🔝9953056974🔝
 
OS-operating systems- ch04 (Threads) ...
OS-operating systems- ch04 (Threads) ...OS-operating systems- ch04 (Threads) ...
OS-operating systems- ch04 (Threads) ...
 
How to Configure Email Server in Odoo 17
How to Configure Email Server in Odoo 17How to Configure Email Server in Odoo 17
How to Configure Email Server in Odoo 17
 
DATA STRUCTURE AND ALGORITHM for beginners
DATA STRUCTURE AND ALGORITHM for beginnersDATA STRUCTURE AND ALGORITHM for beginners
DATA STRUCTURE AND ALGORITHM for beginners
 
Presiding Officer Training module 2024 lok sabha elections
Presiding Officer Training module 2024 lok sabha electionsPresiding Officer Training module 2024 lok sabha elections
Presiding Officer Training module 2024 lok sabha elections
 
Introduction to AI in Higher Education_draft.pptx
Introduction to AI in Higher Education_draft.pptxIntroduction to AI in Higher Education_draft.pptx
Introduction to AI in Higher Education_draft.pptx
 
History Class XII Ch. 3 Kinship, Caste and Class (1).pptx
History Class XII Ch. 3 Kinship, Caste and Class (1).pptxHistory Class XII Ch. 3 Kinship, Caste and Class (1).pptx
History Class XII Ch. 3 Kinship, Caste and Class (1).pptx
 
Biting mechanism of poisonous snakes.pdf
Biting mechanism of poisonous snakes.pdfBiting mechanism of poisonous snakes.pdf
Biting mechanism of poisonous snakes.pdf
 

Process chemistry AS PER PCI SYLLABUS FOR M.PHARM

  • 1. Pharmaceutical Process Chemistry SHIKHA D. POPALI HARSHPAL SINGH WAHI GURUNANAK COLLEGE OF PHARMACY
  • 2. INTRODUCTION 2 “Drug” –a compound that interacts with a biological molecule, triggering a physiological effect. Drugs (active pharmaceutical ingredients) can be classified into 4 groups depending on their origin; 1) Natural products 2) Fermentation products 3) Semi-synthetics –substances produced by partial synthesis 4) Completely synthetic products
  • 3. Background  Average cost of developing a drug was $500 million in 1999 and takes 12-25 years usually.  The chances of a candidate drug that is identified becoming approved is approximately 1 in 10,000. (For every 10,000 trial compounds, 20 reach trials on animals, 10 reach clinical trials on humans and 1 gets FDA approval)  Discovery stage (research labs) involves preparation of potential drug compound on 10 mg to 10 g scale. Toxicology tests are carried out on this material. If these are successful, greater quantities of the compound will be required for clinical trials.  At this stage, development work begins. -Immediate goal – to produce clinical trial material. -Longer term goal– to develop a commercially viable scaled-up validated process. 3
  • 4. Terminology  Development–covers all work between research and production (e.g. analytical, chemical, formulation) and continues when production begins.  Scale-up– process of going from laboratory preparation to whatever scale of manufacture is required to satisfy the market demand (usually 1,000 to 50,000 L range)  Technology Transfer -Transfer between manufacturing sites -Transfer within a manufacturing site (Partnerships for collaborative R & D) 4
  • 5. Process chemistry 5 Process chemistry involves development of practical, safe and cost effective processes for the synthesis of compounds selected to progress from research/discovery to a larger scale. Within the pharmaceutical industry synthetic routes developed within medicinal chemistry often have to be completely redesigned by process chemists, and this is in part due to their differing requirements. The differences between medicinal and process chemistry and the multitude of factors that need to be taken into consideration when scaling up a chemical process.
  • 6. Medicinal chemistry Vs  For medicinal chemists diversity and flexibility of a route is important.  They synthesise a wide range of analogues of a lead compound on a small scale (20 mg) for testing in order to increase activity, reduce side effects and to produce an API that may be easily and efficiently administered to the patient.  For this the compound must have the right properties in terms of activity, toxicity, solubility, pharmacokinetics and pharmacodynamics. 6  In contrast, process chemistry involves development of practical, safe and cost effective processes for the synthesis of compounds on a larger scale (kg to several tonnes) that have been selected to progress from medicinal chemistry.  They generally therefore work on a single target molecule and define the best route to that target. Process chemistry
  • 7. Process chemistry 7 The key stages in process chemistry for the development of an active pharmaceutical ingredient
  • 8. Process chemistry 8 The key stages in process chemistry for the development of an active pharmaceutical ingredient
  • 9. Process chemistry 9 The key stages in process chemistry for the development of an active pharmaceutical ingredient
  • 10. DOE in Process Parameters 10 With the most recent ICH and FDA guidances endorsing a new paradigm of process validation based more on process understanding and control of parameters and less on product testing. After using process knowledgeto relate the attributes to each process unit operation, the inputs and outputs of each unit operation are defined to determine process parameters and in- process controls.
  • 11. DOE in Process Parameters 11 ICH Q8(R2)provides thefollowingdefinitionsusing thetermcritical: • Critical process parameter (CPP).A processparameterwhosevariabilityhasanimpactona criticalquality attributeand,therefore,shouldbemonitoredorcontrolledtoensurethe processproducesthe desired quality. • Critical qualityattribute (CQA).A physical,chemical, biological,or microbiological propertyor characteristicthatshouldbewithin anappropriatelimit, range,or distributiontoensurethedesired product quality. The definition for CPP states that a parameter is considered critical when its variability has an impact on a CQA. The amount of impact is not defined, which leads to the question, does even a small impact to a CQA mean that the parameter is critical?
  • 12. DOE in Process Parameters 12 It is not difficult to imagine the example of an extreme shift of a process parameter having a minor impact on a CQA, whether measurable or not. Extreme temperatures can destroy many pharmaceutical products; however, if a process inherently cannot produce such temperatures, is temperature still considered to be critical and, therefore, required to be monitored and controlled?
  • 13. DOE in Process Parameters 13 When these definitions are strictly interpreted, then one of two extremes would be: • Every quality attribute is critical (they all ensure product quality); every parameter is critical (product cannot be made without controlling them) • No parameter is critical because if they are controlled, all quality attributes will pass specifications. Reality lies somewhere between these extremes. Logic and common sense dictate that additional criteria must be necessary to aid in determining criticality. There is great value in understanding not only if a parameter/attribute is critical (i.e., has an impact), but also how much impact the parameter/attribute has.
  • 14. Process chemistry 14 The key stages in process chemistry for the development of an active pharmaceutical ingredient
  • 15. Process chemistry 15 The key stages in process chemistry for the development of an active pharmaceutical ingredient
  • 16. Process chemistry 16 The key stages in process chemistry for the development of an active pharmaceutical ingredient
  • 17. Term synthesis means in Greek “put together”. Synthetic chemistry is the “art” of building-up complex molecular structures of compounds putting together smaller, easily accessible compounds. 17Synthetic Strategy
  • 18. Synthetic Strategy 18 The construction of a complex organic structure, defined target molecule, requires first of all the identification of the smaller fragments that can be used to build-up the final target. In the case of oligomers such as peptides, oligosaccharides or oligonucleotides, the choice is obvious: the constituent monomers are the building blocks, and the synthesis requires the junction of those monomers by condensation. Two functional groups, one for each monomer, are involved in the reaction
  • 19. What is Development?  Broad range- between research and production- and overlaps with both  In API (Active Pharmaceutical Ingredient) manufacture, begins when active substance is identified and activity demonstrated and more active substance is required.  Once a process that works on plant scale is produced and is validated, development work still continues to improve cost, efficiency, quality and environmental impact and to deal with changing circumstances (vendors, equipment, new regulations (FDA21 CFR part11, Risk-Based Approach etc)) 19
  • 20. Development Stages 1) Chemical Development  Begins when activity of potential API is demonstrated and more active substance is required (for clinical trials).  Process research; –New synthetic routes (literature survey) –Some initial optimisation –Yield improvements –Possibly scale-up to large lab equipment/ kilo lab (up to about 20 L) 20
  • 21. Development Stages cont’d 2) Process Development -Optimization (change conditions and parameters) -Minor change of route / intermediate -Cheaper / more efficient reagents (new to market) -Environmentally-friendly reagents and effluent considerations -Yield / concentration improvement -Statistical methods (e.g. Experimental Design; want maximum amount of unbiased information about factors affecting a process from as few observations as possible. Caution -“Facts are stubborn things. Statistics are more pliable”) 21
  • 22. Development Stages cont’d  Process Development cont’d -SCALE-UP (Kilo Lab, Pilot plant trials) -Transfer to Manufacturing Parallel synthesis reactor block -for optimization of condition 22
  • 23. Development Stages cont’d 3) Process Support  Further optimisation (continuous improvement)  Fine-tuning of yield and throughput  Cost reduction  Troubleshooting (reworks, reprocessing, deviations)  New vendor approval (use tests)  Waste minimisation (recycling) 23
  • 24. Chemical Development Selection of Synthetic Route  GET THE BEST ROUTE FROM THE BEGINNING -Difficult to change later  Route should be short, efficient, robust and give a high yield prior to scale-up  Discovery route often not the best. Expedient not optimal. (raw materials may not be available in bulk, process may not be efficient and may be safety hazards on large scale)  Compare like with like during selection (Same level of optimisation and same scale. Are products of same purity obtained? What is the cost comparison?)  Main constraint is TIME 24
  • 25. Chemical Development Selection of Synthetic Route cont’d  Shortest route usually best (less effluent, shorter lead time, short plant occupation, less analytical work)  A convergent synthesis will be cheaper than a divergent (linear) one with the same number of steps  Literature not always correct and perseverance with a reaction is required  Better to try routes with high chance of success but those which offer significant benefits should be attempted even if little precedent 25
  • 26. Chemical Development In Convergent Synthesis, sections of the target molecule are prepared and joined together to form the target molecule. A better yield than a linear synthesis with the same number of steps will be obtained. The advantage is significant if the subroutes converge close to the end of the synthesis. (page 489, “Medicinal Chemistry, An Introduction”, G. Thomas). 26
  • 27. Process Development Optimisation of Synthetic Route –Once Selected  Work up very important  Can steps easily be combined (telescoping)?  Need excellent process control and understanding of process limits (“stress” the reaction –higher temperature, longer reaction time, less efficient mixing). “Critical” steps / parameters (Quality)  Good analytical methods (standards)  Isolate and characterise by-products  A very innocuous process change can often have a significant influence 27
  • 28. Costing of Processes  Must be a standard costing procedure  Should be dated and updated  Assumptions should be stated  May be several factors leading to variation –scale, plant configuration etc.  Compare like with like and be conservative  Carried out by independent person  Want to be able to see which factors increase cost significantly and why 28
  • 29. Scale up process Pharmaceutical Process Scale-Up deals with a subject both fascinating and vitally important for the pharmaceutical industry—the procedures of transferring the results of R&D obtained on laboratory scale to the pilot plant and finally to production scale. 29
  • 30. Why Scale-up? Scale up is basically needed for:  Market growth  Introduction of new processes  Reduction in making expensive errors in design and operation  Concentrate on addressing areas of doubts and uncertainty  Economic feasibility 30
  • 31. Scale-up Product and process development for scaling up is typically move in small steps.  For instance, the development is initially from lab scale to bench scale then move to pilot scale and finally to commercialization scale.  By performing scale up step by step, the risk with large investments could be lessen. 31 Several Steps
  • 32. Application of the SELECT criteria to scale up  Safety  Environmental  Legal  Economic  Control  Throughput 32
  • 33. Application of the SELECT criteria Safety Safety is the number one criteria for research and development. It describes safety risks including thermal and reactive hazards, and toxicity, which become increasingly important as processes are scaled up. 33
  • 34. Application of the SELECT criteriaEnvironmental The pharmaceutical industry follows the environmental ideals of preventing, minimising and rendering harmless any reagents used and wastes generated from processes. Different metrics are utilised within the industry to track and minimise environmental impact. Steps taken to improve environmental credentials for a process are considered 34
  • 35. Application of the SELECT criteriaLegal It describes legal considerations including regulation of substances by governments, environmental legislation and patent law. 35
  • 36. Application of the SELECT criteria Economic It discusses economic considerations. 36
  • 37. Application of the SELECT criteriaControl Quality control is essential for commercial production of an API (active pharmaceutical ingredient). Selectivity, stability, purification, registration and validation of processes, product specifications and genotoxic impurities all need to be considered to ensure quality is suitable for market. 37
  • 38. Application of the SELECT criteriaThroughput Making sure that we can produce enough within a given time, where chemical yields plays an important role, trying to work as concentrated as possible; if you have one very diluted step in the middle of synthesis this can really be a bottleneck. If you have to perform that several steps before the API this might be real bottleneck and not allow you to make a sufficient volume. What's quite often used is a telescoping of reactions, meaning you work in the same solvent for a number of consecutive steps which is really improving throughput because -you don't need to isolate intermediates, -don't need to filter or centrifugate them, -you don't need to dry them as -you don't need to bring them again or dissolve them again for the next step So, that's one way to really improve throughput.. 38
  • 39. Summary: Application of the SELECT criteria 39 Criteria Sub-criteria Examples of Potential Issues Safety Process safety Explosions Exposure to harmful substances Carcinogens, sensitisers Environmental Wasted resources Quantity & variety of solvents Substances harmful to the environment Aquatic toxins, ozone depleters Legal Infringement of Intellectual Property Competitor patenting key intermediate Regulation of reagents and intermediates Economic Meeting Cost of Good (profit margin) Long synthesis or expensive starting materials Investment costs (equipment) Control Control of quality Meeting specifications/ GMP Control of physical/chemical parameters Throughput Time scale of manufacture Long synthetic route Availability of starting materials Rare natural products
  • 41. 41 Stages of Scale up process
  • 42. Planning for Scale up  Decide on process  Decide on batch size (not too large an increase)  Order raw materials (allow for 10% lower yield)  Carry out safety tests (Exotherms, HAZOP –hazard and operability studies)  Discuss the process and plant requirements with production manager/engineer (materials of construction). Existing multipurpose plant usually.  Prepare safety data sheets and discuss handling of hazardous reagents or intermediates  Ensure analytical procedures, equipment and staff are available 42
  • 43. Planning for Scale up cont’d Write out detailed procedure (assume nothing);  Cleaning check and preparative work  Charging and weighing  Temperature and time limits  Sampling (in-process checks)  Transfers  Work-up and isolation  Drying and effluent disposal/treatment Make allowance for delays / problems (rework procedures, identify suitable “hold points”) 43
  • 44. Planning for Scale up cont’d Leave space in procedure to record data and observations Highlight safety procedures and steps to take if spillage occurs Provide training 44
  • 45. Planning for Scale up cont’d Significant Differences Between Lab and Plant  Heat transfer  Agitation (frothing)  Mass transfer (affects kinetics)  Visibility –of reactions, separations, for cleanliness checks  Separation (stirring, not shaking)  Time (slower additon rates and heating/cooling times, longer work up)  Hazards (Toxic, Exotherm and Electrostatic)  Off-gas treatment  Safe and efficient sampling techniques needed (Process Analytical Technology -Real time reaction monitoring; FT-IR, Particle size, etc.) 45
  • 46. Planning for Scale up cont’d Process Development Taking Account of Differences Between Lab and Plant Equipment  Use safety data already produced to help characterise reaction  Study effect of scale-dependent factors (mixing, mass transfer and heat transfer) at lab scale  Plan a logical set of experiments in time-frame available  Mimic plant conditions; slower addition of reagents, slower rate of heat increase and decrease and mixing, lab system should be baffled if reactors are (no vortex)  Obtain mass and heat transfer data for reactor type, agitator type and solvents to be used.  Use simulation software to predict effects of mass and heat transfer and mixing changes on scale-up. 46
  • 47. Essential steps 47 Laboratory Mock-UPS (Source: Euzen et al., 1993)
  • 48. Production Capacity Table below shows the general production capacity of each scaling up step in the process industries. 48 Scaling factor Typical production capacity Bench/ Laboratory 0.001 – 0.1 kg/h Pilot Plant 10 – 100 kg/h Demonstration Plant 100 – 1000 kg/h Commercial Plant > 1000 kg/h
  • 50.  The collection and evaluation of data, from the process design stage through commercial production, which establishes scientific evidence that a process is capable of consistently delivering quality products. (FDA)  Documented evidence which provides a high degree of assurance that a specific process will consistently result in a product that meets predetermined specifications and quality characteristics. (WHO)  The documented evidence that the process, operated within established parameters, can perform effectively and reproducibly to produce a medicinal product meeting its predetermined specifications and quality attributes. 50 Validation of Large scale Process
  • 51. Validation of Large scale Process Traditional vs new paradigm Post approval changes/chan ge controls/risk analysis Development- Basic Process validation- 3 batches Pilot batch manufacturing Enhanced- Development and process qualification Control Strategy ICH Q8, QbD Continuous and extensive monitoring of CQAs and CPPs for each production batch ICH Q9 and Q10 51
  • 52. 52 Pre-validation phase Protocol Preparation Information from product development studies (identification of critical attributes) Information from primary/ clinical manufacturing (scale up information) Process risk assessment information (identification of critical steps) Validation phase Protocol execution Includes demonstration of content uniformity of the clinical batch Process validation phases Post valdn phase: Review of process, deviations, failures, need for improvement, scale up etc…
  • 53. FDA, January 2011 WHO, Revised Annex 7 of WHO GMP guide (draft for comment) European Medicines Agency (EMA), February 2014 Continuous process verification (CPV) Continuous process verification (CPV) Alternative approaches: -Traditional approach -Continuous process verification -Hybrid approach Process design and Initial validation (process qualification- PPQ) are initial phases of CPV Process design and initial validation (initial process verification) are initial phases of CPV CPV protocol to be supported by extensive dev. information and lab or pilot scale data. Executed on each production batch No mention of number of batches for initial process performance qualification / validation (rather must be justified based on overall product and process understanding) Mentions data on at least three pilot or production batches collected as part of process design Number of batches specified for traditional approach - minimum of three production batches unless other wise justified 53 Latest guidelines
  • 54. Prospective validation Concurrent validation Retrospective validation Protocol reviewed and accepted, Product PQD; OR Protocol executed before submission or PQ Protocol reviewed and accepted, Product PQD Protocol does not need to be submitted Execute and finalize process validation on the first three production batches Execute and finalize process validation on the first three production batches Prepare product quality review report on already manufactured production batches Commercial batches to be released only after satisfactorily conclusion of process validation on three batches Each validation batch can be validated and released. Applicable for low demand products (such as NTDs, orphan drugs or other seasonal products) Applicable for submissions meeting criteria for established products as described in Annex 4, TRS 970 54 Types of process validation and dossier requirements
  • 55.  Design Qualification( DQ) verification process to meet particular requirement relating to the quality of Pharmaceutical and manufacturing process. 55 Process validation- Role of assessment Design qualificat ion Operatio nal qualificati on Performa nce qualificati on Process validatio n GMP Dossier  Performance qualification (PQ) (process qualification)- process of testing to ensure that the individual and combined systems function to meet agreed performance criteria on a consistent basis and to check how the result of testing is recorded. The purpose is to ensure that the criteria specified can be achieved on a reliable basis over a period of time.  Operational Qualification (OQ) process of testing to ensure individual and combined systems function to meet agreed performance criteria and to check how the result of testing is recorded. The purpose is to ensure that all the dynamic attributes comply with the original design.  DQ plan covers user requirement, user specification, Technical specification and DQ report.  Process Validation :- Owners are responsible for Validating Their Processes (personnel, equipment, methods, SOPs) to ensure compliance to cGMP/GLP regulations.
  • 56. Case study; Process Development for Labetalol Production BACKGROUND  Labetalol –antihypertensive, 30,000 Kg p.a. produced by Schering-Plough. Labetalol Manufacturing Process  Mono-pot reaction in jacketed, glass-lined 10,000 L reactor.  Addition of liquid reagents and jacket temperature computer controlled. Solid reagents charged manually via handwhole.  Phase separation using sight-glass.  Solid product from second step isolated by centrifugation.  Product tested to determine if recrystallisation necessary.  When required purity achieved, product is dried 56
  • 57. Labetalol Process –Step 1  Solvent system modified to isopropanol / ethyl acetate containing hydrogen bromide from methanol / ethyl acetate to reduce formation of third impurity. Original process used chloroform.  Concentration was increased threefold increasing throughput and reducing solvent waste. 57 Main Impurities
  • 58. Labetalol Process – Step 2 Process Development  Large excess of dibenzylamine used to ensure reaction driven to completion (cheap reagent and easily washed out)  Propylene oxide added as it reacts with HBr side-product as it’s produced. Presence of HBr would neutralise dibenzylamine and no reaction to give product would occur. Propylene bromohydrin side-product easily washed out.  Use gentle reflux to ensure propylene oxide doesn’t escape 58 +
  • 59. Summary Stages of development are Chemical Development, Process Development and Process Support  Development essential before scale-up and validation –get process right first  Time constraints a major factor  Have process in control (“critical” parameters)  Consider differences between lab and plant scale. Try to mimic plant conditions in lab to anticipate effects when scale up.  Prepare process description carefully for scale-up  Development always on-going 59

Editor's Notes

  1. Will cover syntheticaspects of pharmaceutical production processes. Average cost of developing a drug was $500 million in 1999 and takes 12-25 years usually. The chances of a candidate drug that is identified becoming approved is approximately 1 in 10,000. (For every 10,000 trial compounds, 20 reach trials on animals, 10 reach clinical trials on humans and 1 gets FDA approval) Discovery stage (researchlabs) involves preparation of potential drug compound on 10 mg to 10 g scale. Toxicology tests are carried out on this material. If these are successful, greater quantities of the compound will be required for clinical trials. At this stage, development work begins. -Immediate goal–to produce clinical trial material. -Longer term goal–to develop a commercially viable scaled-up validated process.
  2. Synthetic Strategy The construction of a complex organic structure, defined target molecule, requires first of all the identification of the smaller fragments that can be used to build-up the final target. In the case of oligomers such as peptides, oligosaccharides or oligonucleotides, the choice is obvious: the constituent monomers are the building blocks, and the synthesis requires the junction of those monomers by condensation. Two functional groups, one for each monomer, are involved in the reaction
  3. Pharmaceutical Process Scale-Up deals with a subject both fascinating and vitally important for the pharmaceutical industry—the procedures of transferring the results of R&D obtained on laboratory scale to the pilot plant and finally to roduction scale. The primary objective of the text is to provide insight into the practical aspects of process scale-up. In mixing applications, scale-up is indeed concerned with increasing the linear dimensions from the laboratory to the plant size. On the other hand, processes exist (e.g., tableting) for which “scale-up” simply means enlarging the output by increasing the speed. To complete the picture, one should point out special procedures (especially in biotechnology) in which an increase of the scale is counterproductive and “scale-down” is required to improve the quality of the product.
  4. Scale is answer the question How to scale up from small scale to large scale? Product and process development for scaling up is typically move in small steps. • For instance, the development is initially from lab scale to bench scale then move to pilot scale and finally to commercialization scale. • By performing scale up step by step, the risk with large investments could be lessen.
  5. So the first criterion is Safety. No coincidence that this is really the number one criteria because we cannot afford to develop a process which on large scales is failing and that there is, for example, an explosion, where potentially people can die and that also there can be a complete destruction of the commercial supply chain, where also it could mean that patients do no longer have access to the medicine at a certain moment. There are at least two types of, or two categories of hazards. The first one is thermal and reactive hazards, has to do with the property of the reaction itself, to avoid that there is a thermal runaway we have the reaction mixture is getting to a temperature above which it is no longer stable. Second, on the large scale if there is a reaction where gas is evolving. It's completely different than on lab scale. If you are doing, for example, a BOC de-protection on small-scale, you do not worry about the fact that you are generating iso-butene, but on large scale we are talking about potentially significant volumes of iso-butene during the BOC de-protection. That's just one example, but at de-methylation with HBr where you are generating methyl bromide, you might not consider that in a lab environment but on large scale it's an important attention point. Certainly potential explosives, to be careful when handling these on large scale. Some of the reagents we are using are really corrosive, so on the long term we have to make sure that we can run, we can keep our equipment in a decent state. So we have to select the right equipment and protect it as much as possible. And also pyrophoric material, pyrophoric reagents, need special attention; for example, for storage, but also for dosing, for neutralization after the reaction . The second category are the toxic hazards, where the general principle is that first of all we try to avoid them. Avoid them by not using them, by using other reagents which are less toxic. If there is no real alternative, we try to reduce them; reduce the amount and after that we try to control that. On the one hand by containment and by engineering the process and the equipment, such that it's in a closed environment. And it's really needing also to protect the workers who has to handle these reagents on a large scale. And there's a difference between acute and chronic toxicity as you are aware of, for some of them you will observe it immediately that there's something wrong, for others you only see the effect after a long time. General principle is: If you can't scale it safely, don't scale it at all. A little bit of foam in the lab can be funny, not a problem, but some foaming on large scale, uncontrolled, and it can be, potentially, very toxic what's coming out of the reactor here. I'm sure you agree with me that you don't want to to see that..
  6. So the first criterion is Safety. No coincidence that this is really the number one criteria because we cannot afford to develop a process which on large scales is failing and that there is, for example, an explosion, where potentially people can die and that also there can be a complete destruction of the commercial supply chain, where also it could mean that patients do no longer have access to the medicine at a certain moment. There are at least two types of, or two categories of hazards. The first one is thermal and reactive hazards, has to do with the property of the reaction itself, to avoid that there is a thermal runaway we have the reaction mixture is getting to a temperature above which it is no longer stable. Second, on the large scale if there is a reaction where gas is evolving. It's completely different than on lab scale. If you are doing, for example, a BOC de-protection on small-scale, you do not worry about the fact that you are generating iso-butene, but on large scale we are talking about potentially significant volumes of iso-butene during the BOC de-protection. That's just one example, but at de-methylation with HBr where you are generating methyl bromide, you might not consider that in a lab environment but on large scale it's an important attention point. Certainly potential explosives, to be careful when handling these on large scale. Some of the reagents we are using are really corrosive, so on the long term we have to make sure that we can run, we can keep our equipment in a decent state. So we have to select the right equipment and protect it as much as possible. And also pyrophoric material, pyrophoric reagents, need special attention; for example, for storage, but also for dosing, for neutralization after the reaction . The second category are the toxic hazards, where the general principle is that first of all we try to avoid them. Avoid them by not using them, by using other reagents which are less toxic. If there is no real alternative, we try to reduce them; reduce the amount and after that we try to control that. On the one hand by containment and by engineering the process and the equipment, such that it's in a closed environment. And it's really needing also to protect the workers who has to handle these reagents on a large scale. And there's a difference between acute and chronic toxicity as you are aware of, for some of them you will observe it immediately that there's something wrong, for others you only see the effect after a long time. General principle is: If you can't scale it safely, don't scale it at all. A little bit of foam in the lab can be funny, not a problem, but some foaming on large scale, uncontrolled, and it can be, potentially, very toxic what's coming out of the reactor here. I'm sure you agree with me that you don't want to to see that..
  7. So the first criterion is Safety. No coincidence that this is really the number one criteria because we cannot afford to develop a process which on large scales is failing and that there is, for example, an explosion, where potentially people can die and that also there can be a complete destruction of the commercial supply chain, where also it could mean that patients do no longer have access to the medicine at a certain moment. There are at least two types of, or two categories of hazards. The first one is thermal and reactive hazards, has to do with the property of the reaction itself, to avoid that there is a thermal runaway we have the reaction mixture is getting to a temperature above which it is no longer stable. Second, on the large scale if there is a reaction where gas is evolving. It's completely different than on lab scale. If you are doing, for example, a BOC de-protection on small-scale, you do not worry about the fact that you are generating iso-butene, but on large scale we are talking about potentially significant volumes of iso-butene during the BOC de-protection. That's just one example, but at de-methylation with HBr where you are generating methyl bromide, you might not consider that in a lab environment but on large scale it's an important attention point. Certainly potential explosives, to be careful when handling these on large scale. Some of the reagents we are using are really corrosive, so on the long term we have to make sure that we can run, we can keep our equipment in a decent state. So we have to select the right equipment and protect it as much as possible. And also pyrophoric material, pyrophoric reagents, need special attention; for example, for storage, but also for dosing, for neutralization after the reaction . The second category are the toxic hazards, where the general principle is that first of all we try to avoid them. Avoid them by not using them, by using other reagents which are less toxic. If there is no real alternative, we try to reduce them; reduce the amount and after that we try to control that. On the one hand by containment and by engineering the process and the equipment, such that it's in a closed environment. And it's really needing also to protect the workers who has to handle these reagents on a large scale. And there's a difference between acute and chronic toxicity as you are aware of, for some of them you will observe it immediately that there's something wrong, for others you only see the effect after a long time. General principle is: If you can't scale it safely, don't scale it at all. A little bit of foam in the lab can be funny, not a problem, but some foaming on large scale, uncontrolled, and it can be, potentially, very toxic what's coming out of the reactor here. I'm sure you agree with me that you don't want to to see that..
  8. So the first criterion is Safety. No coincidence that this is really the number one criteria because we cannot afford to develop a process which on large scales is failing and that there is, for example, an explosion, where potentially people can die and that also there can be a complete destruction of the commercial supply chain, where also it could mean that patients do no longer have access to the medicine at a certain moment. There are at least two types of, or two categories of hazards. The first one is thermal and reactive hazards, has to do with the property of the reaction itself, to avoid that there is a thermal runaway we have the reaction mixture is getting to a temperature above which it is no longer stable. Second, on the large scale if there is a reaction where gas is evolving. It's completely different than on lab scale. If you are doing, for example, a BOC de-protection on small-scale, you do not worry about the fact that you are generating iso-butene, but on large scale we are talking about potentially significant volumes of iso-butene during the BOC de-protection. That's just one example, but at de-methylation with HBr where you are generating methyl bromide, you might not consider that in a lab environment but on large scale it's an important attention point. Certainly potential explosives, to be careful when handling these on large scale. Some of the reagents we are using are really corrosive, so on the long term we have to make sure that we can run, we can keep our equipment in a decent state. So we have to select the right equipment and protect it as much as possible. And also pyrophoric material, pyrophoric reagents, need special attention; for example, for storage, but also for dosing, for neutralization after the reaction . The second category are the toxic hazards, where the general principle is that first of all we try to avoid them. Avoid them by not using them, by using other reagents which are less toxic. If there is no real alternative, we try to reduce them; reduce the amount and after that we try to control that. On the one hand by containment and by engineering the process and the equipment, such that it's in a closed environment. And it's really needing also to protect the workers who has to handle these reagents on a large scale. And there's a difference between acute and chronic toxicity as you are aware of, for some of them you will observe it immediately that there's something wrong, for others you only see the effect after a long time. General principle is: If you can't scale it safely, don't scale it at all. A little bit of foam in the lab can be funny, not a problem, but some foaming on large scale, uncontrolled, and it can be, potentially, very toxic what's coming out of the reactor here. I'm sure you agree with me that you don't want to to see that..
  9. So the first criterion is Safety. No coincidence that this is really the number one criteria because we cannot afford to develop a process which on large scales is failing and that there is, for example, an explosion, where potentially people can die and that also there can be a complete destruction of the commercial supply chain, where also it could mean that patients do no longer have access to the medicine at a certain moment. There are at least two types of, or two categories of hazards. The first one is thermal and reactive hazards, has to do with the property of the reaction itself, to avoid that there is a thermal runaway we have the reaction mixture is getting to a temperature above which it is no longer stable. Second, on the large scale if there is a reaction where gas is evolving. It's completely different than on lab scale. If you are doing, for example, a BOC de-protection on small-scale, you do not worry about the fact that you are generating iso-butene, but on large scale we are talking about potentially significant volumes of iso-butene during the BOC de-protection. That's just one example, but at de-methylation with HBr where you are generating methyl bromide, you might not consider that in a lab environment but on large scale it's an important attention point. Certainly potential explosives, to be careful when handling these on large scale. Some of the reagents we are using are really corrosive, so on the long term we have to make sure that we can run, we can keep our equipment in a decent state. So we have to select the right equipment and protect it as much as possible. And also pyrophoric material, pyrophoric reagents, need special attention; for example, for storage, but also for dosing, for neutralization after the reaction . The second category are the toxic hazards, where the general principle is that first of all we try to avoid them. Avoid them by not using them, by using other reagents which are less toxic. If there is no real alternative, we try to reduce them; reduce the amount and after that we try to control that. On the one hand by containment and by engineering the process and the equipment, such that it's in a closed environment. And it's really needing also to protect the workers who has to handle these reagents on a large scale. And there's a difference between acute and chronic toxicity as you are aware of, for some of them you will observe it immediately that there's something wrong, for others you only see the effect after a long time. General principle is: If you can't scale it safely, don't scale it at all. A little bit of foam in the lab can be funny, not a problem, but some foaming on large scale, uncontrolled, and it can be, potentially, very toxic what's coming out of the reactor here. I'm sure you agree with me that you don't want to to see that..
  10. Finally, the last criteria which is important is the Throughput. Making sure that we can produce enough within a given time, where chemical yields plays an important role, trying to work as concentrated as possible; if you have one very diluted step in the middle of synthesis this can really be a bottleneck. So, for example, certain types of chemistry like metathesis, ring-closing metathesis, which is in most cases run at very diluted conditions. If you have to perform that several steps before the API this might be real bottleneck and not allow you to make a sufficient volume. What's quite often used is a telescoping of reactions, meaning you work in the same solvent for a number of consecutive steps which is really improving throughput because you don't need to isolate intermediates, don't need to filter or centrifugate them, you don't need to dry them as you don't need to bring them again or dissolve them again for the next step so that's one way to really improve throughput..
  11. Finally, the last criteria which is important is the Throughput. Making sure that we can produce enough within a given time, where chemical yields plays an important role, trying to work as concentrated as possible; if you have one very diluted step in the middle of synthesis this can really be a bottleneck. So, for example, certain types of chemistry like metathesis, ring-closing metathesis, which is in most cases run at very diluted conditions. If you have to perform that several steps before the API this might be real bottleneck and not allow you to make a sufficient volume. What's quite often used is a telescoping of reactions, meaning you work in the same solvent for a number of consecutive steps which is really improving throughput because you don't need to isolate intermediates, don't need to filter or centrifugate them, you don't need to dry them as you don't need to bring them again or dissolve them again for the next step so that's one way to really improve throughput..
  12. Bench scale and lab scale systems are important early-stage tools for assessing and scaling new technology. Application are a precursor to larger pilot and demonstration scale plants. Researchers can then plot reaction yield and selectivity under a variety of operating conditions( Temp. Pressure Change solvent and volume.) Reactor volumes in bench and lab scale systems are typically less than 1000ml. The most common operating configurations at the bench or lab scale are batch for continuous stirred tank reactor (CSTR), or autoclave etc. Pilot plants Scale provide the first window into continuous processing and often incorporate unreacted feed or product recycle. They are the workhorse of the process development world. Catalyst performance tests are carried out to determine, or confirm, yield and selectivity data, and the lifetime of the catalyst is measured under a variety of operating conditions. Reactor size at the pilot plant scale typically ranges from 1 and 100 litres. Demonstration Scale differ from pilot plants in that the equipment and process flowsheet much more closely resemble commercial scale operations. Extended operating runs permit catalyst lifetime studies over a longer period of time, while significant quantities of final product can be generated for market testing. Developers need to go to through the demonstration scale to prove to the market and investors that their technology meets its performance expectations, such as product yield and properties, and catalyst life, and is ready for commercialization.
  13. Bench scale and lab scale systems are important early-stage tools for assessing and scaling new technology. Application are a precursor to larger pilot and demonstration scale plants. Researchers can then plot reaction yield and selectivity under a variety of operating conditions( Temp. Pressure Change solvent and volume.) Reactor volumes in bench and lab scale systems are typically less than 1000ml. The most common operating configurations at the bench or lab scale are batch for continuous stirred tank reactor (CSTR), or autoclave etc. Pilot plants Scale provide the first window into continuous processing and often incorporate unreacted feed or product recycle. They are the workhorse of the process development world. Catalyst performance tests are carried out to determine, or confirm, yield and selectivity data, and the lifetime of the catalyst is measured under a variety of operating conditions. Reactor size at the pilot plant scale typically ranges from 1 and 100 litres. Demonstration Scale differ from pilot plants in that the equipment and process flowsheet much more closely resemble commercial scale operations. Extended operating runs permit catalyst lifetime studies over a longer period of time, while significant quantities of final product can be generated for market testing. Developers need to go to through the demonstration scale to prove to the market and investors that their technology meets its performance expectations, such as product yield and properties, and catalyst life, and is ready for commercialization.
  14. Pharmaceutical Process Scale-Up deals with a subject both fascinating and vitally important for the pharmaceutical industry—the procedures of transferring the results of R&D obtained on laboratory scale to the pilot plant and finally to roduction scale. The primary objective of the text is to provide insight into the practical aspects of process scale-up. In mixing applications, scale-up is indeed concerned with increasing the linear dimensions from the laboratory to the plant size. On the other hand, processes exist (e.g., tableting) for which “scale-up” simply means enlarging the output by increasing the speed. To complete the picture, one should point out special procedures (especially in biotechnology) in which an increase of the scale is counterproductive and “scale-down” is required to improve the quality of the product.
  15. Pharmaceutical Process Scale-Up deals with a subject both fascinating and vitally important for the pharmaceutical industry—the procedures of transferring the results of R&D obtained on laboratory scale to the pilot plant and finally to roduction scale. The primary objective of the text is to provide insight into the practical aspects of process scale-up. In mixing applications, scale-up is indeed concerned with increasing the linear dimensions from the laboratory to the plant size. On the other hand, processes exist (e.g., tableting) for which “scale-up” simply means enlarging the output by increasing the speed. To complete the picture, one should point out special procedures (especially in biotechnology) in which an increase of the scale is counterproductive and “scale-down” is required to improve the quality of the product.
  16. Pharmaceutical Process Scale-Up deals with a subject both fascinating and vitally important for the pharmaceutical industry—the procedures of transferring the results of R&D obtained on laboratory scale to the pilot plant and finally to roduction scale. The primary objective of the text is to provide insight into the practical aspects of process scale-up. In mixing applications, scale-up is indeed concerned with increasing the linear dimensions from the laboratory to the plant size. On the other hand, processes exist (e.g., tableting) for which “scale-up” simply means enlarging the output by increasing the speed. To complete the picture, one should point out special procedures (especially in biotechnology) in which an increase of the scale is counterproductive and “scale-down” is required to improve the quality of the product.
  17. Pharmaceutical Process Scale-Up deals with a subject both fascinating and vitally important for the pharmaceutical industry—the procedures of transferring the results of R&D obtained on laboratory scale to the pilot plant and finally to roduction scale. The primary objective of the text is to provide insight into the practical aspects of process scale-up. In mixing applications, scale-up is indeed concerned with increasing the linear dimensions from the laboratory to the plant size. On the other hand, processes exist (e.g., tableting) for which “scale-up” simply means enlarging the output by increasing the speed. To complete the picture, one should point out special procedures (especially in biotechnology) in which an increase of the scale is counterproductive and “scale-down” is required to improve the quality of the product.
  18. Pharmaceutical Process Scale-Up deals with a subject both fascinating and vitally important for the pharmaceutical industry—the procedures of transferring the results of R&D obtained on laboratory scale to the pilot plant and finally to roduction scale. The primary objective of the text is to provide insight into the practical aspects of process scale-up. In mixing applications, scale-up is indeed concerned with increasing the linear dimensions from the laboratory to the plant size. On the other hand, processes exist (e.g., tableting) for which “scale-up” simply means enlarging the output by increasing the speed. To complete the picture, one should point out special procedures (especially in biotechnology) in which an increase of the scale is counterproductive and “scale-down” is required to improve the quality of the product.
  19. The collection and evaluation of data, from the process design stage through commercial production, which establishes scientific evidence that a process is capable of consistently delivering quality products. (FDA) Documented evidence which provides a high degree of assurance that a specific process will consistently result in a product that meets predetermined specifications and quality characteristics. (WHO) The documented evidence that the process, operated within established parameters, can perform effectively and reproducibly to produce a medicinal product meeting its predetermined specifications and quality attributes.(EMA)
  20. The collection and evaluation of data, from the process design stage through commercial production, which establishes scientific evidence that a process is capable of consistently delivering quality products. (FDA) Documented evidence which provides a high degree of assurance that a specific process will consistently result in a product that meets predetermined specifications and quality characteristics. (WHO) The documented evidence that the process, operated within established parameters, can perform effectively and reproducibly to produce a medicinal product meeting its predetermined specifications and quality attributes.(EMA)
  21. Design Qualification( DQ) is a verification process on the design to meet particular requirement relating to the quality of Pharmaceutical and manufacturing process. DQ plan covers user requirement, user specification, Technical specification and DQ report. Operational Qualification (OQ) is the process of testing to ensure that the individual and combined systems function to meet agreed performance criteria and to check how the result of testing is recorded. The purpose is to ensure that all the dynamic attributes comply with the original design. Performance qualification (PQ), also called process qualification, is the process of testing to ensure that the individual and combined systems function to meet agreed performance criteria on a consistent basis and to check how the result of testing is recorded. The purpose is to ensure that the criteria specified can be achieved on a reliable basis over a period of time. Process Validation :- Owners are responsible for Validating Their Processes (personnel, equipment, methods, SOPs) to ensure compliance to cGMP/GLP regulations.
  22. Solvent system modified to isopropanol / ethyl acetate containing hydrogen bromide from methanol / ethyl acetate to reduce formation of third impurity. Original process used chloroform. Concentration was increased threefold increasing throughput and reducing solvent waste.
  23. Labetalol Process –Step 2, Process Development Large excess of dibenzylamine used to ensure reaction driven to completion (cheap reagent and easily washed out) Propylene oxide added as it reacts with HBr side-product as it’s produced. Presence of HBr would neutralise dibenzylamine and no reaction to give product would occur. Propylene bromohydrin side-product easily washed out. Use gentle reflux to ensure propylene oxide doesn’t escape