CE70007 - Pharmaceutical Development Flashcards

1
Q

What is route selection?

A

Route Selection refers to the selection of starting materials, process intermediates, and general chemistry conditions associated with the route, but not necessarily the final “optimised” operating conditions.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

What is the manufacturing process description?

A

The Manufacturing Process Description is the defined process that must be adhered to during manufacturing. It is part of the regulatory submission, and every aspect must be backed-up with data.

A good manufacturing process:
• Provides DS that routinely meets CQAs and MAs
• Embraces the principles of inherent safety
• Is cost effective
• Is supported by accurate and reproducible analytical methods
• Consists of well understood process operations and provides the right level of control
• Embraces the principles of green chemistry and environmental sustainability

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What’s CQA, CPP, and MA?

A

Critical Quality Attribute (CQA)
Critical Process Parameter (CPP)
Manufacturing Attribute (MA)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What are the main stages of pharmaceutical R&D?

A
  1. Lead ID (lab stage)
  2. Pre-clinical
  3. Phase I (small pilot)
  4. Phase II (small-full pilot)
  5. Phase III (full pilot)
  6. File and launch
  7. Lifecycle management (manufacturing)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What’s CGS?

A

Cost of goods

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What’s CMC?

A

Chemistry, Manufacturing & Control

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What is GMP?

A

Good Manufacturing Practice
It is that part of Quality Assurance which ensures that medicinal products are consistently produced and controlled to the quality standards appropriate to their intended use.

It applies to: people, products, procedures, processes, and premises.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Why work to GMP?
Good manufacturing practise

A

Any Pharmaceutical Company has
• to ensure patient safety
• to work to best practice standards to produce quality products fit for purpose
• to provide confidence to the prescribers and patients
• to sell products

“Fit for Purpose” Pharmaceuticals
• Right product
• Right purity/strength
• Free from contamination
• No deterioration
• Right packaging/labelling
• Properly sealed and protected against
damage and contamination
• Complies with all legal & regulatory requirements

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What’s the qualification process for a new pharmaceutical design?

A
  1. User requirements specification
  2. Validation master plan
  3. Design qualification
  4. Purchase order
  5. Installation qualification
  6. Start-up
  7. Operational qualification
  8. Routine operation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What’s process validation?

A

Process validation is the collection and evaluation of data, which establishes scientific evidence that a process is capable of consistently delivering quality product.

Process validation does not strictly apply during development, but it does in manufacturing.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Define POL, SOP, EOP, GUI

A

POL: policies. High level outline of intent

SOP: standard operating procedure

EOP: Equipment operating procedures

GUI: Guidelines / Best Practise

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What are the objectives of the FDA?

A

FDA objectives:
– Promote new technology
– Introduce modern quality management techniques
– Risk-based approaches to R&D and inspection
– Promote science-based policies
– Improve FDA drug quality regulatory programs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What’s an API?

A

Active pharmaceutical ingredient

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What is PAT?

A

Process Analytical Technology - a means of analysing and monitoring a process using off-line, at-line and on-line techniques.

It has been developed and used in many areas including the food and water industries and petrochemicals in particular.

Recent advances in measurement (spectrometers) and processing (data analysis) speeds has made the approach more amenable to real-time analysis.

The FDA encouraged the Pharmaceutical Industry to utilise PAT in 2003/4

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What is QbD?

A

Quality by Design.

A systematic approach to development that begins with predefined objectives and emphasises product and process understanding and process control, based on sound science and quality risk management.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Give examples of PAT instruments:

A

• Near Infrared NIR
• Mid IR
• UltraViolet / Visible
• Raman
• Lasentec (FBRM)
• Mass Spec

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What is FBRM?

A

Focused Beam Reflectance Measurement

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

How does Focused Beam Reflectance Measurement (FBRM) work?

A

A solid-state laser light source provides a continuous beam of monochromatic light that is launched down FBRM® probe. A set of lenses focuses the laser light to a small spot. This focal spot is carefully calibrated to be positioned at the interface between the probe window and the actual process. Tightly controlling the position of the focal spot is necessary for a sensitive and repeatable measurement.
A precision motor is used to rotate the optics at a constant speed. The speed is monitored and controlled throughout the measurement to ensure maximum precision in the data. Standard probes operate to provide a fixed 2 m/s scan speed.
The focused beam scans a circular path at the interface between the probe window and the particle system. As the beam sweeps across the face of the probe window, individual particles or particle structures will backscatter the laser light back to the probe.

These pulses of backscattered light are detected by the probe and translated into Chord Lengths based on the simple calculation of the scan speed (velocity) multiplied by the pulse width (time); a chord length is simply defined as the straight-line distance from one edge of a particle or particle structure to another edge.

Thousands of individual chord lengths are typically measured each second to produce the Chord Length Distribution which is the fundamental measurement provided by FBRM®.

Note that unlike other particle size analysis techniques, with FBRM® measurement there is no assumption of particle shape

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

How are UV/Vis and NIR fibres formed?

A

UV/Vis (Ultraviolet/Visible) and NIR (Near-Infrared) fibres are typically made by drawing molten silica or other materials into thin strands. The process involves heating and stretching a glass preform to create long, flexible fibres.

Pure silica and surrounding the core is a doped-fluorine silica cladding. A polyimide material is then applied. Then a coiled stainless steel jacketing is applied over the core, cladding and buffer to protect the fibre and provide strain relief.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What are the 2 main types of mid-IR fibres?

A

Chalcogenide glasses composed of sulfur (S), selenium (Se), and tellurium (Te) with the addition of other elements such as germanium (Ge), arsenic (As), and antimony (Sb) that result in the formation of stable glasses.
Typical melt temperatures range from 600°C to 900°C, depending upon composition.

Silver halide solid solution crystals - AgCl : AgBr

(Typical fibre lengths:
NIR - 1000 m+
IR - 2-3 m
UV - 5m )

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What is SAM-Spec?

A

SAM-Spec ® is a technique based on Spatially Resolved Spectroscopy
combined with several chemometric approaches. The system uses :

– A light source for irradiation
– Measurements are performed at several distances for physical characterisation
– Absorbance Spectral measurements provide chemical characterisation
– Symmetrical acquisition positions provide homogeneity evaluations.

[Technique from video on analysing tableting]

  1. NIR is injected into the media and will immediately be isotropically (in all directions) scattered into the media.
  2. Photons propagate at different depths. Their density decreases while receding from light source (i.e. greater density of photons nearer where light was incident).
  3. NIR chemical spectra are recorded by the probe at several depths and locations simultaneously.
  4. SAM-spec measures up to 12 spectra at once to provide an accurate reading (of tablet conc.). This can address issues in homogeneity and any coatings used. The more dense the material, the greater the degree of scattering.
  5. Info on spatial distances gives information on hardness, density, porosity, and particle size.
  6. Cracks and deformities interfere with normal scattering.
  7. All this takes place in about 5 ms.

26 measurement points and 10-40 images per tablet

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What PAT techniques are suitable to monitor crystallisation?

A

UV
Mid IR
Sonic velocity
NIR
FBRM

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

Which technique is most suitable for monitoring distillation?

A

NIR, monitoring distillate solvent composition
Mid IR

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What PAT technique is suitable for monitoring tableting?

A

NIR imaging

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

What PAT technique is suitable for monitoring dryers?

A

NIR
Mass spec
Miniature spectrometers.

Using a linear variable filter the MicroNIR, detector, light source, collection optics, and electronics are fully integrated in one small USB-powered palm-size device weighing <60 grams. Available wavelength range is 950 – 1650 nm.

Can be attached to:
•a rotary blender
•agitated dryer
to determine the end-point

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

What is PCA?

A

Principal component analysis

PCA is for the analysis (exploration) of a single data set (matrix)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

What PAT methods are suitable for monitoring reactions?

A

UV
NIR
Mid-IR
Mass spec

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

What’s PLS?

A

Partial least squares

A PLS model (or calibration) is built taking spectra of known well characterised materials. Spectra acquired during the process in real-time are then compared to the model to predict component concentrations.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

What are the types of PAT process interfaces?

A

Direct via dip pipe, by-pass loop

Probe
- ATR
- Transmission
- Reflectance

Flow cell

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

Define control strategy:

A

A planned set of controls, derived from current product and process understanding, that assures process performance and product quality. The controls can include parameters and attributes related to drug substance and drug product materials and components, facility and equipment operating conditions, in-process controls, finished product specifications, and the associated methods and frequency of monitoring and control.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

Define critical quality attribute (CQA):

A

A physical, chemical, biological or microbiological property or characteristic that should be within an appropriate limit, range, or distribution to ensure the desired product quality.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

Define critical process parameter:

A

A process parameter whose variability has an impact on a critical quality attribute and therefore should be monitored or controlled to ensure the process produces the desired quality.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

Define edge of failure:

A

The boundary to a variable or parameter, beyond which the relevant quality attributes or specification cannot be met.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

What does pharmaceutical development include?

A

• Defining the target product profile as it relates to quality, safety and efficacy, considering e.g., the route of administration, dosage form, bioavailability, dosage, and stability

• Identifying critical quality attributes (CQAs) of the drug product, so that those product characteristics having an impact on product quality can be studied and controlled

• Determining the quality attributes of the drug substance, excipients etc., and selecting the type and amount of excipients to deliver drug product of the desired quality

• Selecting an appropriate manufacturing process

• Identifying a control strategy

• Identifying, through e.g., prior knowledge, experimentation, and risk assessment, the material attributes and process parameters that can have an effect on product CQAs

• Determining the functional relationships that link material attributes and process parameters to product CQAs

• Using the enhanced process understanding in combination with quality risk management to establish an appropriate control strategy which can, for example, include a proposal for design space(s) and/or real-time relea

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

Describe the QbD (quality by design) approach:

A

Step 1 - Identify the desired profile of the marketable product ie the Target Product Profile and the CQA’s of the product.

Step 2 - Selection of the synthesis route for the API (Active Pharmaceutical Ingredient) and processing requirements for the formulation type, together with the associated unit operations required for these processes. (Suitable unit operations include for example, Chemical Reaction, Crystallisation, Distillation, Drying, Blending and Tableting.)

Step 3 - Gaining experimental knowledge by operating the process and exploring the effect of options and changes.
Refining and optimising the process to determine the Proven Acceptable Ranges (PAR’s).

Step 4 - Develop the process Control Strategy including Design Space definitions.

Step 5 - Technology transfer of the process from Development to the Manufacturing business.

Step 6 - Manufacture of marketable product and Regulatory Submission.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

What does CQA stand for?

A

Critical Quality Attribute (CQA): A physical, chemical, biological or microbiological property or characteristic that should be within an appropriate limit, range, or distribution to ensure the desired product quality.

Each CQA has an individual control strategy with associated elements of control to ensure that all of the CQAs are satisfied to meet the required patient safety and efficacy criteria.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

What’s a design space?

A

The multidimensional combination and interaction of input variables (e.g., material attributes) and process parameters that have been demonstrated to provide assurance of quality.

The way input attributes and process parameters affect the output quality attributes must be described through a quantitative model which permits an assessment of the level of assurance of quality for any multi-dimensional combination of input attributes and process parameters.

First Principle (Mechanistic) Approach
– A first principle, or mechanistic, model is a combination of experimental data and mechanistic knowledge of chemistry, physics, and engineering which enables the prediction of process performance. A first principle, or mechanistic model, is derived from an understanding of the underlying science e.g. chemical reaction kinetics.

Empirical Modelling Approach
– Carefully planned experimentation incorporating DoE studies (an efficient method for determining the impact of multiple parameters and their interactions) is used to obtain data which are used to derive both the form of the model and the associated unknown model coeffi

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

How is a parallel coordinates plot shown?

When is it used?

A

In the parallel coordinates plot, a set of equally spaced parallel axes are drawn for each variable (eg batch descriptor: CQA, QCPP, End-point, etc). Then a given row of data (eg values for a single batch) is represented by drawing a line that connects the values of that row on each corresponding axis.

Parallel Coordinates allow multidimensional data to be visualised

  1. Representation of Design Space
    • Proven Acceptable (and Normal Operating) Ranges
    can be displayed for single or multiple unit operations.
  2. Representation of Continuous Verification.
    • Verification that a process Control Strategy
    maintains operation within the Design Space limits can be illustrated
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

What are the elements of control?

A

A control strategy comprises 3 possible modes of control

• Attribute control - testing to specification ie. starting materials, reagents, intermediates, drug substance etc using on-line or off-line methods.

• Parametric control
– Individual PAR limits applied to process parameters.
– Interactive PAR model based (Design Space relationships) limits
applied to process parameters.

• Procedural Control - order of addition, work-up procedure, extraction wash sequences etc

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

How does process validation play a part in 3 different stages of design?

A

Stage 1 – Process Design: The commercial process is defined during this stage based on knowledge gained through development and scale-up activities.

• Stage 2 – Process Qualification: During this stage, the process design is confirmed as being capable of reproducible commercial manufacturing.

• Stage 3 – Continued Process Verification: Ongoing assurance is gained during routine production that the process remains in a state of control.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

Where is process verification applied?

A

It can be applied in 2 distinct phases :
• Achieving the point of commercialisation. Before release of batches for commercial supply, a manufacturer is expected to have accumulated enough data and knowledge about the production process to support post approval distribution.

• During the production phase, continual verification or monitoring will allow evaluation of quality indicator data, changes and adverse trends. This will be performed periodically to decide if new studies or other verification experiments need to be done. This demonstrates that the process continues to work as intended and remains in control

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

What are the 3 golden rules of mixing

A

A deep vortex is not a sign of good mixing

Impeller speed rarely stays constant on scale up

Mixing performance is inversely proportional to scale

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

What to T, D, H, H’, C, and B refer to regarding standard vessel geometry?

A

T - tank diameter
D - baffle / stirrer diameter
B - baffle width
H - liquid level height
H’ - tank base height
C - impeller off-bottom clearance

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

What does partial baffles refer to?

A

A tank having 3 or less baffles

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

Explain axial and radial flow from mixers:

A

Axial:
Fluid flows up and down the vessel
- low shear
- good for suspended solids

Radial:
Propeller blades are pitched, so fluid will go sideways, hit the vessel wall, and move up and down vessel, circling in a figure of 8 manner
- high shear
- good for dispersions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

When are axial and radial mixers / impellers used?

A

Axial:
- low shear purposes
- good for suspended solids

Radial:
- high shear purposes
- good for dispersions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

What is the static volume?

A

The tank volume at the bottom of the tank below the impeller where nothing moves.

Roughly 1% nominal tank volume.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

What’s the minimum stir volume?

A

Min volume needed for impeller to stir fluid. Is useful for cleaning.

Roughly 5% nominal tank volume.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

What’s the minimum mixed volume?

A

This determines the minimum batch sized vessel.

It is about 30-40% the nominal tank volume, and is the volume required for effective mixing to take place.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

What is the issue with conical bottom shaped tanks?

A

They are very bad mixers.

They make good separators.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

What are the main impeller types?

A

Radial flow impeller
• Discharges liquid radially outwards towards vessel walls

Axial flow impeller
• Discharges liquid axially towards base or liquid surface depending on rotation direction

Mixed flow impeller
• Flow predominantly in axial direction with also a radial component

Close clearance impeller (e.g. anchor impeller)
• Ensures good motion
near vessel walls

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

What are the process applications of radial flow impellers?

A
  • turbulent and transitional regime
  • gas-liquid
  • liquid-liquid dispersions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

What are the process applications of axial and mixed flow impellers?

A
  • turbulent and transitional regime
  • blending
  • solid suspension
  • liquid-liquid dispersions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

What are the process applications of close clearance impellers?

A
  • laminar regime
  • blending
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

What are the predominant baffle types?

A

The beaver tail baffle - oval/cylindrical with a flat end.
Can be glass lined, but is not very effective

Cylindrical baffles with additional cylindrical protrusions.

C-shaped baffles

Beavertail and finger baffles are often used in glass-lined vessels
– Smooth shape suitable for glassing

Other internals can provide a degree of baffling
– Dip pipes
– Heating/cooling coils

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

What may a mixing process be controlled by?

A

A process may be controlled by one or more of:

Liquid blending
• reaction
• homogenisation

Solid-liquid mixing
• solid catalysed reaction
• dispersion

Gas-liquid mixing
• fermentation
• hydrogenation

Dispersing immiscible liquid
• reaction
• emulsions

Heat transfer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

How is power consumption calculated from torque?

A

P=2 π N Λ

Where Λ is torque and N is rotational speed.

P and Λ determine the capital and running costs
– If the impeller must start from rest in a bed of solids, the power and torque requirements for start-up can be extremely large, and depend on the characteristics of the solid.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

How is torque measured?

A

– Small scale measurements
• Mount vessel or motor on frictionless bearings and measure torque using a load cell or dynamometer
• Use a commercial torque meter
• Use a modified rheometer

– At larger scales:
• Strain gauges can be used.
• Electrical current can be used.
– Subject to many errors
– Estimate of losses in the gearbox and bearings required

Beware of:
– Vibration & resonance problems

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

What does power draw refer to?

A

The power drawn by an impeller

P = Po ρ N^3 D^5

Where:
Po - power number
Rho - fluid density
N - impeller rotational speed
D - impeller diameter

Po, or Newton number, Ne – Depends on
• Impeller type
• Impeller and vessel dimensions
• Properties of the phases present

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

How can flow regime be determined in a stirred tank considering the power number?

A

Re < 10 - laminar flow
Po proportional to 1/Re

10 < Re < 10^3 - transitional flow
Po = f(Re)

Re > 10^3 - turbulent
Po = constant

Fully turbulent, Re»10^4

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

What is blend time?

A

Time taken to reach 95% homogeneity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

What is just suspension speed?

A

Speed at which no particle stays stationary for more than one to two seconds at the bottom of the vessel.

(Njs)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

List key drug administration routes:

A
  • oral (tablets, capsules, liquids)
  • pulmonary (aerosols, inhalation powders)
  • transdermal (creams, controlled-release adhesive patches)
  • intra-venous (e.g. vaccines, insulin, antibiotics)
  • “direct application” (e.g. eye drops, skin cream)
  • implantable controlled-release devices, depot systems…
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

What does Tmax refer to regarding drug delivery?

A

Time until blood-drug conc. is at its highest.
With a single dose, there is an increase and decrease in blood-drug conc. Meanwhile, with chronic administration, the concentration (fluctuates but) increases and plateaus over time.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

What’s the therapeutic index?

A

Difference between minimum toxic concentration and minimum effective concentration.

The maximum blood-drug concentration must remain between these two concentrations.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

What are MTC and MEC with regards to drug delivery?

A

Minimum toxic concentration and minimum effective concentration

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

What does LADME stand for, with regards to drug delivery?

A

Liberation
Absorption
Distribution
Elimination (Biotransformation/metabolism and excretion)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

What is bioavailability?

A

Bioavailability F = the fraction of the administered dose that reaches
the systemic circulation as the parent drug (not as metabolites)

F = AUC / Dose

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

What is apparent volume of distribution?

A

V = Total amount of drug in the body / Plasma concentration

It is important when considering hydrophobic drugs/particles.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

What is the partition coefficient?

A

A measure of lipophilic character of drug.

A partition coefficient is the ratio of the concentration of a substance in one medium or phase (C1) to the concentration in a second phase (C2) when the two concentrations are at equilibrium; that is, partition coefficient = (C1/C2)equil.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

What does the Noyes Whitney equation show?

A

Dissolution rate of particles.

The Noyes Whitney equation,

dC/dT = kA(Cs-Ct)

describes the change in concentration over time (dC/dT) as a function of the dissolution rate constant k, surface area A, solubility Cs, and concentration in the bulk fluid Ct.

k may be replaced by D/δ = diffusivity / apparent thickness

dm/dt = -D/δA(C*-C.bulk)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

Key questions for pharmaceutical design?

A

What’s being made

How much active ingredient and in what form?

What are the requirements on dissolution / release kinetics?

Overall product composition?

Process route?

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

What are the different particle milling techniques?

A

Stirred media mils (wet milling)
Impact/Jet mills (micronisation)
Attrition mills

Dry milling however is not suitable when trying to obtain very small particle sizes, due to electrostatic forces.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

What’s the mixing number, regarding powder blending?

A

Nmix - number of unit mixing operations required for the system to reach a given state of mixedness

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

What’s segregation, regarding powder blending?

A

The natural tendency of powders to de-mix due to difference
in particle size, shape, density or surface properties (friction, cohesion)

  • occurs during transport (conveying) or storage (IBCs) of powders
  • need to “freeze” a well-mixed state immediately after blending
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

Why coat tablets?

A

-Taste masking
-Visual appeal (coloured tablets)
-Protective layer (abrasion)
-Delayed release effect (gastric fluid resistance)
-Functional coating of carrier particles - API

Typical film thickness: 5-50 um

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

Describe the principle of wet granulation:

A

Contact powder with a liquid binder, wet powder particles become cohesive, agglomeration occurs during particle collisions, binder sets to form mechanically stable granules.

Binder types:
- melt binders ~ melts, solidify upon cooling (e.g. PEG)
- aqueous binders ~ solutions, solidify upon drying (e.g. HPC, PVP)
- water ~ partial dissolution and recrystallisation of ingredient(s)

Binder application:
- spray (liquid atomisation) for low-shear processes
- mechanical dispersion in high-shear processes

Granulation processes:
- fluid bed granulation
- high-shear mixer granulation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

What is involved in primary and secondary manufacturing?

A

Primary - API manufacturing, reaction route selection, separations and solids isolation (crystallisation/filtration/drying)

Secondary - blending, forming, types of dosage routes, processing routes, no chemical transformation taking place, mechanical unit operations

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

What are exipients?

A

A constituent of a medicine other than the active substance, added in the formulation for a specific purpose.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

What are binders (in medicine)?

A

Binding agent or binders are employed to convey the cohesiveness to the granules. Binders are added to the tablet formulation to impart plasticity as well as increases inter-particulate bonding strength in the tablet that ensure the tablet remains intact after compression.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

What are the 4 classes of the Biopharmaceutics Classification System (BCS)?

A

I) High solubility & high permeability (dissolve well and absorb fast)

II) Low solubility & high permeability (dissolution rate needs to be improved during production to enhance absorption)

III) High solubility & low permeability (rate limiting step of drug is the absorption across intestinal wall. High API conc is needed to increase driving force / supersaturating the conc’ in the intestine)

IV) Low solubility & low permeability (as above, rate limiting step is absorption across intestine wall)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

What are typical components/’ingredients’ of a tablet?

A

API / Active pharmaceutical ingredient

Filler (bulking agent)

Binder

Disintegrant or matrix-forming polymer

Lubricant / glidant

Coating polymer

Pigment

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

Why would particles need to be milled during drug development?

A

To reduce particle size and improve / increase the API dissolution rate.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

Describe wet milling:

A

Wet milling, also known as wet grinding, is a process through which particles that are suspended in a liquid slurry are dispersed in that liquid by shearing or crushing. Once the milling process is complete, these particles are ready for use or can be dried and separated for incorporation into additional products.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
85
Q

What quality attributes are considered for tablets?

A

Hardness
Attrition
Weight uniformity
Content uniformity
Disintegration time
Release kinetics

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
86
Q

What are the main steps of the overall crystallisation process design procedure?

A

1) Thermodynamics (Solid-liquid equilibria, looking at solubility)
2) Kinetics (rate of crystallisation)
3) Selection of crystalliser type
- mode of achieving super-saturation
4) Mass, energy and population balances
- to determine operating condition that meet
product specification

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

What’s polymorphism?

A

The ability to crystallize in more than
one crystallographic form

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

What are the 2 types of polymorph?

A

Enantiotropic - can convert into one another / interconvertible (e.g. by T). Possess points at which their solubilities at a given temperature are the same, and can then change form.

Monotropic - incapable of transformation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
89
Q

What’s drowning-out crystallisation?

A

In drowning-out crystallization, supersaturation is generated by adding an anti-solvent, which reduces the solubility of the solute.

This method is used for highly soluble materials rather than evaporative or cooling crystallization, which have weak solubility temperature dependence

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
90
Q

What’s a eutectic system?

A

A eutectic system is a homogenous, solid mixture of two or more substances that form a super-lattice that melts or solidifies at a temperature lower than any of the individual ingredients’ melting point. The term is most usually used to describe a mix of metals.

It melts or solidifies at a temperature lower than the melting point of any one of the constituent components.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
91
Q

Explain congruent melting point:

A

Congruent melting point - hydrate (or solvate) can exist in equilibrium with a solution of the same composition (point D).

92
Q

What are the labile and metastable regions of crystallisation?

A

Labile zone - where nucleation occurs

Metastable zone - growth range

In the metastable region nuclei will develop into crystals, but no nucleation will occur. In the labile region both might be expected to occur.

The final region, at very high supersaturation, is denoted the precipitation region, where this result might be most probable.

93
Q

How is supersaturation created?

A

Creating supersaturation:
- by concentration (evaporation of solvent)
- by temperature (cooling)
- by adding antisolvent

Supersaturation is the driving force for crystallisation.

94
Q

When do primary and secondary nucleation occur?

A

Primary - in the absence of crystals
- homogeneous nucleation = clusters of atoms spontaneously form in the solution
- heterogeneous nucleation = impurities (e.g. dust particles) act as nucleation centres

Secondary - in presence of crystals (seeding)
- contact nucleation mechanism (crystal-crystal and crystal-wall/impeller collisions)
- shear nucleation mechanism (fluid flow over surface creates new nuclei)

We want secondary nucleation over primary as it gives more control over the particles that are formed.

95
Q

What’s crystal habit?

A

The term crystal habit describes the favoured growth pattern of the crystals of a mineral species, whether individually or in aggregate.

96
Q

Why is the volume fraction of crystals in slurries aimed to be kept around 15%?

A

Prevent the slurry from becoming too thick.

97
Q

How is particle size distribution often considered?

A

Plot of particle size vs number of particles.
Normal distribution expected.

98
Q

What is a MSMPR crystalliser and its features?

A

Mixed-suspension mixed-product removal crystalliser

  • ideally mixed
  • steady-state, continuous operation
  • no crystals in feed
  • no agglomeration or breakage
  • size independent growth rate
99
Q

What are issues to be considered during filtration?

A

Be careful about the following:
- Particle breakage in agitated filters
(resuspension)
- Cake washing: acceptable product loss vs. product purity
- Cake deliquoring: shrinkage/cracking of cake
- Filtration centrifuge: uniformity of cake thickness

100
Q

What are issues to be considered during drying process design and operation?

A

Be careful about the following:
- Geometrical similarity during scale-up
(surface/volume ratio)
- Rate-limiting step: heat vs. mass transfer
- Particle agglomeration/breakage in agitated dryers
- Phase changes in crystals (solvates/hydrates)

101
Q

What parameters effect the performance of spray drying processes?

A

Related to particle size and morphology…
- Feed conc
- Droplet size / atomisation pressure
- Solvent choice
- Additive choice

Related to product temperature and residual moisture…
- Feed flowrate
- Drying gas flowrate
- Drying gas humidity
- Drying gas temperature

102
Q

Describe the two stages of single droplet drying:

A

1st period - Evaporation from free surface. Wet bulb temperature.

2nd period - Diffusion across solid shell. Formation of hollow core. .

103
Q

What are the main types of chromatography?

A

Different mechanisms from different combinations of gaseous, solid or liquid phases give rise to the main types of chromatography.
* Adsorption
* Partition
* Ion Exchange
* Size Exclusion

Chromatography is a technique for the separation of a mixture by passing it in solution or suspension through a medium in which the components move at different rates.

104
Q

What does adsorption chromatography involve?

A

It uses a solid stationary phase e.g. silica gel, activated carbon, and a liquid or gaseous mobile phase.

Solutes are separated according to their adsorption characteristics onto the stationary phase.

105
Q

What does partition chromatography involve?

A

It is based on a thin film formed on the surface of a solid support by a liquid stationary phase.

Solutes equilibrate between the mobile and stationary phase

106
Q

What does ion exchange chromatography involve?

A

A resin (stationary solid phase) is used to covalently attach anions or cations onto it.

Solute ions of opposite charge in mobile liquid phase are attracted to the resin by electrostatic forces.

107
Q

What does molecular or size exclusion chromatography involve?

A

Aka gel permeation or gel filtration, it lacks any attractive interaction between the stationary phase and solute.

The liquid or gas phase passes through a porous gel which separates molecules based on their size.

The gel consists of spherical beads with pores of a specific size distribution. The small pores exclude the larger solute molecules, but allow smaller molecules to enter the gel, causing them to be retained.

This causes the larger molecules to pass through the column at a faster rate than the smaller ones.

108
Q

List key chromatography techniques:

A

High-performance liquid chromatography (HPLC or uHPLC)
Gas chromatography
Supercritical fluid (SFC)
Thin layer (TLC)
Capillary electrophoresis
Chiral

109
Q

Describe reverse-phase HPLC:

A

Reverse-phase HPLC involves binding organic molecules to a non-polar stationary phase, often silica derivatized with alkyl chains, in a relatively polar environment (the mobile phase), which could contain water, and then eluting the organic molecule using a gradient of a less polar organic solvent.

The compounds are therefore separated on account of their differing hydrophobic character.

The less polar and more hydrophobic solutes are more strongly retained on a reversed phase column.

110
Q

What is electroosmotic flow?

A

The motion of liquid induced by an applied potential across a porous material, capillary tube, membrane, microchannel, or any other fluid conduit.

This looks at the movement of bulk solution

111
Q

What is electrophoretic mobility?

A

Electrophoretic mobility is the solute’s response to the applied electrical field in which cations move toward the negatively charged cathode, anions move toward the positively charged anode, and neutral species remain stationary.

112
Q

Describe gel electrophoresis:

A

Electrophoresis is a process that enables the sorting of molecules based on size.

Using an electric field, molecules (such as DNA) can be made to move through a gel made of agarose or polyacrylamide.

The electric field consists of a negative charge at one end which pushes the molecules through the gel, and a positive charge at the other end that pulls the molecules through the gel. The molecules being sorted are dispensed into a well in the gel material.

The gel is placed in an electrophoresis chamber, which is then connected to a power source. When the electric field is applied, the larger molecules move more slowly through the gel while the smaller molecules move faster.

The different sized molecules form distinct bands on the gel

113
Q

How does capillary electrophoresis (CE) work?

A

The capillary of the CE system is made of fused silica, having inner negatively charged layers. Thus, when high voltage is supplied at the end of the capillary tube, the molecules in a mixture start separating based on their ionic mobility and interaction with the liquid phase or electrolyte medium.

The molecules or ions with smaller sizes or higher charges move faster in the molecules than those larger in size or having more charges. Additionally, the molecules can also be concentrated by creating a gradient in the pH or conductivity of the electrolyte solution.

The separated molecules are then detected by a detector, which displays them on screen as distinct peaks as a function of migration time. The electrophoretic mobility of molecules in response to an electric field depends on their charge, radius, and solvent viscosity.

114
Q

What is the retention factor, considering chromatography?

A

Compounds will spend some time in the stationary phase and some time in the mobile phase. The time spent by an individual molecule in each of the two phases is called the capacity or retention factor, denoted as ‘k.’ The ratio of time spent in the two phases is equal to the ratio of the mass of the compounds in the two phases.

k = time spent in stationary phase / time in mobile phase

= mass in stationary / mass in mobile

k = (tr-t0)/t0

115
Q

What is the partition coefficient, K?
How is it found?

A

K = molar conc in stationary phase / molar conc in mobile phase

It is the ratio of the conc of a compound in the two phases.

116
Q

What is phase ratio, β?

A

β = volume of mobile phase / volume of stationary phase

k = K / β

Where k is the retention factor and K is the partition coefficient.

117
Q

What’s retention time?

A

The time it takes for a compound to travel through the column (from when an analyte is injected to when it reaches the detector) is known as the retention time (tr). If a compound is not retained at all, it will still take time to travel through the column.
Therefore, to establish a relationship between ‘k’ and retention time, an adjustment must be made to account for this travel time.

118
Q

What is dead time, t0?

A

The time it takes for an unretained compound to travel through the column, t0

119
Q

How is retention factor, k, calculated considering retention time and dead time?

A

k = (tr - t0)/t0

Where t0 is dead time

120
Q

What is the number of theoretical plates, N, with regards to chromatography?

A

Also known as column efficiency, the number of theoretical plates is a mathematical concept and an indirect measure of peak width for a peak at a specific retention time.

Number of Theoretical Plates (N):
N = 5.54 * (tR / wh)^2

Where:
N = number of theoretical plates
tR = retention time
wh = peak width at half height

A column with a high number of theoretical plates will have a narrower peak at a given retention time than a column with a lower N number. High column efficiency is beneficial since it requires less peak separation (meaning lower alpha, α - selectivity) to completely resolve components. On stationary phases where the alphas (α) are small, more efficient columns are needed.

Column efficiency is a function of the column dimensions (diameter, length, and film thickness), the type of carrier gas, its flow rate or average linear velocity, and the compound and its retention. For column comparison purposes, the number of theoretical plates per meter (N/m) is often used.

121
Q

What is the height equivalent to a theoretical plate, H, with regards to chromatography?

A

A measure of column efficiency, usually reported in mm.
The shorter each theoretical plate, the more plates are ‘contained’ in any length of column. This translates to more plates per meter and a higher column efficiency.

Height Equivalent to a Theoretical Plate (H):
H = L / N

Where:
H = Height equivalent to a theoretical plate
N = number of theoretical plates
L = Length of column (mm)

122
Q

How is resolution considered for chromatography when analysing results?

A

The higher the resolution, the less overlap there is between two peaks.

Separation is simply the distance or time between two peak maxima (alpha, α). Resolution takes into consideration both alpha (α - selectivity) and the width of the peaks.

It can be calculated using either of the equations below. Baseline resolution typically occurs at a resolution number of 1.50. However, there might not be a visible baseline between the two peaks. Numbers greater than 1.50 indicate a baseline between the peaks, while numbers less than 1.50 suggest some degree of co-elution.

Theoretical Aspects:

Resolution (R):
R = 1.18 * (tR2 - tR1) / (wh1 + wh2)
or
R = 2 * (tR2 - tR1) / (wb1 + wb2)

Where:
R = Resolution
tR1 = Retention time of the first peak
tR2 = Retention time of the second peak
wh1 = Peak width at half height of the first peak (time)
wh2 = Peak width at half height of the second peak (time)
wb1 = Peak width at base of the first peak (time)
wb2 = Peak width at base of the second peak (time)

123
Q

Considering Height Equivalent to a theoretical plate, H, what does the value of H depend on?

A

The value of H relies primarily on four main factors:

The velocity of the mobile phase.
Multipath diffusion, denoted by constant A, representing diffusion due to packing non-uniformity.
Longitudinal diffusion coefficient in the mobile phase (expressed by constant B), inversely related to the mobile phase’s velocity.
Mass transfer terms, including Cs for the stationary phase and Cm for the mobile phase.
For High-Performance Liquid Chromatography (HPLC) columns packed with particles, these factors are accounted for by the following formula:

H = A + B/u + (Cs+Cm)⋅u

Where:

H represents the height equivalent to a theoretical plate.
u stands for the average linear velocity of the mobile phase.
A is a constant indicating diffusion due to non-uniform packing.
B represents the longitudinal diffusion coefficient in the mobile phase.
Cs and Cm denote the mass transfer terms in the stationary and mobile phases, respectively.

This formula combines these elements to determine the efficiency or height equivalent to a theoretical plate in HPLC columns.

124
Q

For the height of plates equation,

H = A + B/u + (Cs + Cm)*u

Explain the importance of A, B/u, and Cu.

A

A: In packed columns, peak broadening occurs due to various factors. As analyte molecules traverse the column, they follow diverse pathways around the packed particles. Certain pathways are inevitably longer than others, causing the molecules to spread out as they move through the column. The extent of this spreading is influenced by the column material’s nature and the quality of packing. Generally, this factor is directly proportional to the particle size of the packing material. It’s a crucial consideration for packed columns; however, capillary columns, lacking particles, negate this factor.

The Longitudinal Diffusion Term (B/u):
Longitudinal diffusion contributes to peak broadening as analytes diffuse from areas of high concentration to less concentrated areas before and after the moving band. The packing material reduces the extent of longitudinal diffusion to some degree. At low velocities, longitudinal diffusion negatively impacts resolution, although this effect diminishes at higher velocities. Notably, this term holds significant importance in gas chromatography due to significantly higher diffusion coefficients in gases than in liquids. In liquid chromatography, this term is typically near-zero relative to other factors.

The Mass Transfer Terms (Cu):
The mass transfer term highlights the perpetual lack of equilibrium between the mobile and stationary phases within a chromatography column. Analytes in the mobile phase require time to move into the stationary phase, but equilibrium isn’t fully attained. Consequently, some analytes are carried ahead of the main band. Equally, molecules take time to move out of the stationary phase, leaving certain analyte molecules behind due to the swift movement of the mobile phase. While both longitudinal diffusion and mass transfer are diffusion-based, they occur differently concerning flow rate. Longitudinal diffusion occurs parallel to the flow direction, inversely linked to the mobile phase flow rate, while mass transfer diffusion happens perpendicular to the flow rate. Thus, faster mobile phase movement reduces the time available for phase equilibrium, directly impacting peak broadening through the mass transfer effect.

125
Q

What does a thermal conductivity detector (TCD) involve?

A

A Thermal Conductivity Detector (TCD) comprises an electrically heated filament in a temperature-controlled cell. Typically, there is a consistent heat flow from the filament to the detector. However, when an analyte reduces the thermal conductivity of the column effluent, the filament heats up and its resistance changes. This alteration in resistance is detected by a Wheatstone bridge, resulting in a measurable voltage change.

All compounds, regardless of being organic or inorganic, possess a thermal conductivity different from that of helium (the carrier gas).
This detector can sense and detect all compounds.

A TCD exhibits similar responses to comparable concentrations of organic analytes. Hence, it can be used without calibration, allowing estimation of the sample component’s concentration by the ratio of the analyte peak area to all components (peaks) present in the sample.

However, the detector has limitations due to its relatively lower sensitivity in comparison to other detectors. Additionally, it often has a relatively large dead volume, which makes it less suitable for capillary work.

126
Q

What does a flame ionization detector (FID) involve?

A

The Flame Ionization Detector (FID) has nearly universal sensitivity to organic compounds, exhibiting excellent sensitivity and remarkable linearity.

In this detector, the column effluent is directed into a flame fuelled by hydrogen, with forced air flowing.
A potential of several hundred volts is applied between the tip of the flame burner and the collector surrounding the flame.
When sample components combust, they generate a burst of ions, resulting in a current between the flame tip and the collector.

The FID detector offers several advantages. Its response is approximately proportional to the number of carbon atoms present in the flame at any given time.
Moreover, it is insensitive to inorganic gases, water, carbon dioxide, sulphur dioxide, nitrogen oxides, and other non-combustible gases.
Additionally, the detector boasts an extremely wide linear range, spanning about seven orders of magnitude, and has a low dead volume of approximately 1 ml.

127
Q

How can data from chromatograms be quantified?

A

Chromatogram data can be utilized to determine the relative or absolute concentration of components within a mixture, assuming good resolution is achieved.

The peak area, derived from the integration of the detector signal during the elution of a component, is directly proportional to the amount of that particular component in the sample.
However, it’s important to note that a detector’s response can vary among different compounds. For instance, an HPLC ultraviolet detector relies on the absorption of electromagnetic radiation, considering the spectra of the components and the detection wavelength utilized.

To obtain quantitative information, there are four principal methods:
1. Normalizing peak areas
2. Using internal standards
3. Employing external standards
4. Applying standard addition methods.

128
Q

How is chromatography scale up factor calculated?

A

Scale up Factor = (diameter^2 * length)/(Analytical diameter^2 * analytical length)

This factor is used to calculate the preparative column loading.
For example if a 1mg sample loading is used with a 3.9mm ID analytical column, then a 24mg loading is required for a preparative column with a 19mm ID.

Flow rate (prep) = flow rate (analytical) * (prep diameter)^2 / (analytical diameter)^2

129
Q

What are the main focuses of process safety?

A

Preventing injury and death

Preventing damage to plant

Prevent loss of production

Maintain company reputation

130
Q

Why are semi-batch processes preferred in the pharma industry?

A

Easier process control

Rate of reaction can be controlled by the rate of the addition (or removal) of components

Reactive inventory is limited

131
Q

What are the 4 key considerations of process safety?

A
  1. Substitue
  2. Minimise
  3. Moderate
  4. Simplify

Can it be made inherently safer?
– Inherent safety is a philosophy - it seeks to remove hazardous procedures or reagents from the process.

132
Q

What is a key consideration in geometric similarity?

A

Using the single scale ratio, S.

This defines the relative magnitude of all linear dimensions between the large and small scales. It is important that only linear dimensions are considered (e.g. diameter, temperature, height, etc.)

E.g. S = D2/D1

Non-linear parameters e.g. volume can not be considered. Volume would increase by a factor of S^3

133
Q

What does kinematic similarity consider?

A

Velocities at geometrically similar positions remain constant
* Constant tip speed
* Constant superficial gas velocity
* Constant maximum liquid velocity in impeller discharge

134
Q

What does dynamic similarity consider?

A

That the ratio of forces (dimensionless groups) remain constant at different scales
* E.g. constant Froude number for systems in which vortexing for gas entrainment is required

  • Beware:
    – The relationship between process performance and the dimensionless group may not be linear
135
Q

General scale-up advice:

A

Physical operations take longer

Heat transfer is worse

Mixing is more difficult to achieve

Distillations are less efficient

Note - on scale up, impeller speed goes down

136
Q

What are the scale-up rules for the following?

Solid distribution
Gas-liquid
Solid suspension
Liquid blending

A

Solid distribution: equal energy input (P/V)

Gas-liquid: Equal mass transfer kla

Solid suspension: Zwietering (related to mixing)

Liquid blending: Equal tip speed U

137
Q

What is the difference between minimum stirred and minimum mixed volume in a tank?

A

The minimum stirred volume in a tank is the smallest volume at which the impeller is at least partially submerged so that it will provide some fluid motion.
This is suitable for processes where proper mixing is not required, e.g. some basic distillations, or boil-outs for cleaning purposes. This volume is usually less than 10% of the nominal tank volume.

The minimum mixed volume in a tank is the smallest volume at which the agitation system in the tank (impeller(s) & baffles) can create a mixing environment that results in reproducible process performance e.g. for crystallisations. This volume is usually between 30-40% of the nominal tank volume.

When designing a batch plant or a process, it is the minimum mixed volume in a tank that determines the smallest batch size, not the minimum stirred volume.

138
Q

What materials are commonly selected for use in pharmaceutical plants?

A

Common materials of construction for a pharmaceutical plant are (in increasing cost order) glass lined mild steel, stainless steel and hastelloy.

If you need very high corrosion resistance, tantalum is also a possibility but very expensive.

What material you can choose depends on compatibility with your process. If you have to deal with HF (hydrofluoric acid) that would rule out glass (but it may depend on the concentration – if very low, you might be able to get away with special pharmaglass), so you have to check which metal alloys suit.

139
Q

How do you go about choosing a type of crystalliser to use for a solution crystallisation process?

What are the typical things to consider?

A

Primary things to consider are heat transfer and mixing. If you opt for a stirred tank, select a cost effective material of construction that is not only compatible with your process but also has good thermal conductivity.

If you have to use glass-lined steel rather than stainless steel, go for
conductive Pharmaglass and ensure that you choose a heat transfer mode of operation that allows you to keep the required cooling profile easily.

Half coiled heat transfer pipes generate higher U values than traditional baffled jackets and are also easier to maintain.

For the mixing environment, axial flow impellers (e.g. wide blade hydrofoils) are preferable as they exert lower shear and cause less particle size modification; at least three baffles are needed.

You may want to consider multi-flight agitation, if you have a large vessel or have to operate the vessel at different fill volumes.

140
Q

Are there any guidelines about choosing a desired particle size distribution?

A

You should go with the largest mean particle size that the formulators can accept, as large particles mean easier filtrations and shorter plant cycle times.
The acceptable maximum size of API that is included in the tablet is driven by its dissolution and formulation characteristics.
Most APIs get milled before being processed further into tablets, so easy powder flow of the crystals from a hopper into the mill is of prime concern.

Aim for a tight, mono-modal PSD of the API, as this input to the mill produces similar output at the smaller size which is easier to blend with excipients before going to the tablet press.

141
Q

How does a typical pharmaceutical plant handle waste?

A

The waste of a process is separated into organic solvent waste and aqueous waste. Solvent waste gets drummed up and sent to incineration. Most production sites have their own incinerator, while smaller R&D sites send this waste off-site by tanker.

Aqueous waste gets treated in an on-site biological waste processing plant, and a cleaned water stream is returned to the environment.

Waste processing is considered a non-core activity by most pharmaceutical companies and is hence outsourced to specialised contractors.

142
Q

When designing a stirred reactor, is there an upper limit for stirrer speed?

A

The upper speed limit of a stirred vessel is governed by motor size, gearbox size and mechanical stability of the vessel.

It is common to design a reactor with the smallest possible motor, gearbox and stirrer/baffle assembly agreeable with process requirements as this reduces capital and operating costs.

143
Q

How can we obtain a particle size distribution for a crystalliser output, given expressions for the growth and secondary nucleation rate?

How can we quantify the effects of re-dissolving and destroying nuclei, and the suppression of secondary nucleation through control of process variables?

A

If you have an excessively large nucleation rate, then that indicates that you haven’t yet found the right operating conditions for your crystalliser.

In principle, secondary nucleation is caused by collision of crystals with themselves or the internals of the crystalliser.
This in turn is primarily governed by the dissipated energy into the mixture and the supersaturation of the solution.

The dissipated energy into the mixture can be controlled by the rotation speed of the impeller and the type of the mixing system.

To reduce secondary nucleation, select low shear impellers such as wide blade hydrofoils in a fully baffled environment (but eliminate additional reactor inserts such as samplers, coils or plates) and choose slow agitation speeds of larger diameter multi-stage impellers.

Hydrofoils are very effective in creating good circulation in the tank at comparatively low energy input, as they have low Power but high Flow numbers.

Supersaturation can be controlled via the temperature gradient, i.e. a slower cooling profile causes less secondary nucleation.

To quantify all of the above, you have to resort to empirical formulas.
B0 = k(ρa)(εb)(rg)^c

Where:
B0 - nucleation rate
k - system constant
ρ - suspension density
ε - dissipated energy
rg - growth rate

Exponents a, b, c need to be determined experimentally, but typical values are around 1, 0.5-1, 1.3-2.3, respectively.

144
Q

What’s the Penney diagram?

A

Used for plant scale-up processes, the Penney diagram relates the increase of energy input to the scale increase in a double logarithmic plot.

Both quantities are ratios, i.e. (P/V)plant / (P/V)lab and Vplant / Vlab.

If the ratios relate to each other, the same must be true for the individual values, i.e. P/V and V. The volume is essentially a linear dimension cubed (times a constant).

P/V is N cubed D squared times a constant.
Provided that we can express the scale-up rule of ND^m as a factor of N^3D^2, we can work out their corresponding slopes in the double logarithmic plot.

145
Q

What’s the Mechanistic / First Principle approach for design (in pharma)?

A

A first principle, or mechanistic, model is a combination of experimental data and mechanistic knowledge of chemistry, physics, and engineering which enables the prediction of process performance.

A first principle, or mechanistic model, is derived from an understanding of the underlying science e.g. chemical reaction kinetics.

146
Q

What’s the Empirical modelling approach for design (in pharma)?

A

Carefully planned experimentation incorporating DoE [design of experiments] studies (an efficient method for determining the impact of multiple parameters and their interactions) is used to obtain data which are used to derive both the form of the model and the associated unknown model coefficients.

147
Q

What are the different types of HPLC & uHPLC ((ultra)-High performance liquid chromatography?

A

Normal Phase
Reversed Phase
Preparative
Simulated Moving Bed
Ion Exchange
Size Exclusion

148
Q

What are the charges of anions and cations?

A

Anions: Negatively charged

Cations: Positively charged

149
Q

How does normal phase HPLC (high-performance liquid chromatography) work?

A

Polar stationary phase
Non-polar mobile phase

Polar compounds interact more strongly with the polar stationary phase, causing them to move more slowly through the column.
Non-polar compounds interact less and move more quickly. This differential interaction leads to separation of the compounds.

Compounds elute (exit) the column at different times, known as retention times, based on their interactions with the stationary phase.
This allows for identification and quantification by comparing retention times of known standards.

150
Q

How does reversed phase HPLC (high-performance liquid chromatography) work?

A

Non-polar stationary phase
Polar mobile phase

Non-polar compounds interact more strongly with the non-polar stationary phase, causing them to move more slowly through the column.
Polar compounds interact less and move more quickly. This differential interaction leads to separation of the compounds.

Compounds elute (exit) the column at different times, known as retention times, based on their interactions with the stationary phase.
This allows for identification and quantification by comparing retention times of known standards.

Alkyl chains are covalently bonded to the solid support, creating a hydrophobic stationary phase with a stronger affinity for hydrophobic compounds.

151
Q

How does preparative chromatography work?

A

Preparative chromatography is the process of using liquid or supercritical fluids to isolate a sufficient amount of material for other experimental or functional purposes.

The goal of preparative chromatography is to isolate and purify a specific compound or compounds from a mixture.

The choice of column size, stationary phase, and mobile phase depends on the properties of the compounds being separated. (Typically, columns used in preparative chromatography are larger and can handle higher sample loads, with diameters around 2cm - 1m).

The sample containing the mixture to be separated is loaded onto the column (in larger quantities compared to analytical chromatography).
The separation process occurs similarly to analytical chromatography, with compounds interacting with the stationary phase and eluting at different rates based on their affinity for the stationary phase.

As the compounds elute from the column, they’re collected in fractions. These fractions contain the separated compounds and are collected based on their retention times.

152
Q

What’s the difference between analytical and preparative chromatography?

A

Analytical:
- It’s primary goal is to provide information about the composition, concentration, and properties of the substances present in the sample.
- Smaller scale
- Often involves continuous detection and recording of separated components but may not always collect fractions

Preparative:
- Its primary purpose is to obtain larger quantities of pure substances for further use
- Large scale
- Collects fractions containing the separated compounds of interest

153
Q

How does simulated moving bed chromatography work?

A

Continuous counter-current chromatography.

This is accomplished with a series of chromatography columns arranged in a ring. An eluent flow circulates through this ring.

SMB uses a series of switching valves that continuously redirect the flow of the mobile phase and the sample between different zones within the column. These valves control the direction of flow, allowing the stationary phase to behave as if it were moving while actually remaining stationary.

As the mobile phase flows through the column, the components of the mixture interact with the stationary phase. Due to the counter current movement and periodic switching, different components move to different sections of the column, leading to continuous separation.

154
Q

How does ion exchange chromatography (IEC) work?

A

The stationary phase consists of a solid support material with charged functional groups.
For cation exchange chromatography, the stationary phase has negatively charged functional groups (anionic groups like sulfonate or carboxylate).
For anion exchange chromatography, the stationary phase has positively charged functional groups.

When a sample containing ions of interest is introduced to the column, ions with charges opposite to those on the stationary phase are attracted and retained on the stationary phase. Ions with the same charge as the stationary phase move more quickly through the column.

A mobile phase (buffer solution) is passed through the column. By changing the composition or concentration of the mobile phase, the strength of the ionic interactions between the sample ions and the stationary phase can be altered.

As the mobile phase flows through the column, ions are selectively displaced from the stationary phase based on their interactions. The retained ions are released when competing ions in the mobile phase displace them, allowing for separation based on the differences in their affinity for the stationary phase.

The eluted ions are detected using various detectors, such as conductivity, UV-Vis spectrophotometry, or mass spectrometry, to identify and quantify the separated ions.

155
Q

Describe size exclusion chromatography (SEC):

A

The stationary phase in SEC consists of porous beads or a gel matrix. These beads have pores of defined sizes that allow molecules to enter the pores based on their size.

When a sample containing molecules of various sizes is introduced into the column, larger molecules cannot enter the pores of the stationary phase and, therefore, take a shorter path through the column.
Smaller molecules, however, can enter the pores and take a longer, more convoluted path as they move through the column.

A mobile phase (buffer solution) continuously flows through the column, carrying the sample components. As molecules elute from the column, they are detected by a detector.

156
Q

Discuss how packed GC works:

A

Packed column gas chromatography operates using a column filled with a solid support material.
This stationary phase interacts with the sample compounds as they pass through the column.

The sample, typically in a gaseous or vaporized state, is introduced into the column through an injector port. The sample is vaporized and carried by an inert carrier gas through the column.

As the sample travels through the column, different compounds interact differently with the stationary phase based on factors like polarity, size, and volatility.
Compounds with stronger interactions with the stationary phase spend more time in the column.

157
Q

Discuss how capillary GC works:

A

Capillary gas chromatography (GC) is an advanced form of gas chromatography that utilizes a capillary column for separation.

It uses a very narrow, capillary-sized column. These columns are typically coated with a thin layer of stationary phase, often a liquid film or a bonded phase, which provides the separation mechanism.

As the sample components travel through the capillary column, they interact with the stationary phase. Separation occurs based on differences in the interaction of the compounds with the stationary phase, including factors like polarity, size, and volatility.

Capillary GC offers higher resolution, sensitivity, and efficiency compared to packed column GC. It allows for better separation of complex mixtures and can handle smaller sample sizes due to its sensitivity.

158
Q

Describe supercritical fluid chromatography (SFC):

A

SFC typically uses a chromatographic column packed with a stationary phase, often silica-based or other materials designed for compatibility with supercritical fluids.
The mobile phase, usually supercritical CO2, carries the sample through the column.

Separation occurs based on differences in compound interactions with the stationary phase and the supercritical fluid. Compounds with different polarities or sizes interact differently, leading to separation as they move through the column.

159
Q

What’s the Van Deemter plot?

A

A plot of chromatography plate height (H) vs average linear velocity of mobile phase (u).

The van Deemter equation is a hyperbolic function that predicts that there is an optimum velocity at which there will be the minimum variance per unit column length and, thence, a maximum efficiency.

160
Q

Do chromatography columns have plates?
What is the number of theoretical plates for?

A

Chromatography columns don’t have physical plates like those in fractional distillation columns.

The number of theoretical plates is a measure of the efficiency of a chromatography column. It’s a theoretical concept used to describe the effectiveness of separation within the column.
Each theoretical plate represents a hypothetical stage or zone in the column where equilibration between the mobile phase and stationary phase occurs.

The higher the number of theoretical plates, the greater the column’s separation efficiency. A higher number of theoretical plates means better resolution and sharper peaks in the chromatogram.
This implies that compounds spend more time interacting with the stationary phase, resulting in better separation.

The equation used to calculate the number of theoretical plates is the Van Deemter equation, which takes into account different factors influencing the column efficiency, such as the rate of mass transfer, longitudinal diffusion, and eddy diffusion.

161
Q

What’s resolution in chromatography?

How can it be calculated?

A

Resolution refers to the ability of a column or separation method to separate two adjacent peaks or components in a mixture.
It’s a measure of the degree of separation or how well two different analytes are distinguished from each other within a chromatographic system.

For high resolution, these diffusion factors should be minimised (plate height should be small).

R = sqrt(N)/4 * ((a-1)/a) * (k2 / (k2+1))

Where:
k2 is the retention factor of the second (or later eluting) peak
a is selectivity (k2/k2)

(or 2 other existing equations)

162
Q

What’s the effect of particle size on chromatography?

A

Reducing particle size minimizes the impact of flow rate on peak efficiency.

Smaller particles have shorter diffusion paths, facilitating faster movement of solutes in and out of the particles.

The analyte spends less time within the particle, reducing the opportunity for peak diffusion to occur.

As particle size decreases, the Van Deemter curve becomes flatter, showing less susceptibility to higher column flow rates.
Smaller particle sizes result in better overall efficiencies and decreased peak dispersion across a broader spectrum of usable flow rates, albeit with significantly increased back pressures.

163
Q

What is the Giddings relationship for reduced plate height and reduced mobile phase velocity v?

A

h = H / dp
Where:
H is height equivalent of theoretical plate (um)
dp is mean particle size (um)

v = u*dp/Dm
Where:
u is linear velocity of the mobile phase
dp is the particle diameter
Dm is the diffusion coefficient of the solute in the mobile phase

A well-packed column should have a reduced plate height (h) in the range of 2-3 at a reduced velocity (v) of about 3.

With the above parameters, an empirical form of the Van Deemter equation can be derived:

h = B/v + Av^(1/3) + Cv

164
Q

What is a TCD?

A

A thermal conductivity detector

It comprises an electrically heated filament housed in a temperature-controlled cell. In standard conditions, a consistent heat transfer occurs from the filament to the detector body.
As an analyte elutes and decreases the thermal conductivity of the column effluent, the filament experiences a rise in temperature and alters its resistance.
This change in resistance is detected by a Wheatstone bridge, generating a detectable voltage shift.

Given that all compounds, both organic and inorganic, exhibit a thermal conductivity distinct from helium (the carrier gas), the TCD can detect all compounds.
Similar concentrations of organic analytes elicit comparable responses from the TCD, allowing its use without calibration.
Estimation of a sample component’s concentration can be inferred from the ratio of the analyte’s peak area to the total components (peaks) in the sample.

However, the detector’s sensitivity is relatively low compared to other detectors, and it typically possesses a relatively large dead volume. Consequently, it’s not particularly suitable for capillary work.

165
Q

What does normalising peaks areas (chromatography quantitation) involve?

A

It is simply the area of an individual peak calculated as a percentage of the total areas recorded for all peaks in the chromatogram.

166
Q

What does the internal standard method (chromatography quantitation) involve?

A

The internal standard method offers enhanced accuracy in quantitative analysis. It negates the necessity for precisely measured injections by incorporating a reference standard within each analyzed sample. An internal standard is chosen to elute at a strategic point in the chromatogram.

The process involves analyzing a test mixture sample with known quantities of each component and a predetermined amount of the internal standard (I.S.) to compute the Response Factor (RF).

RF = (Ax * Cis)/(Ais * Cx)

Ax is Area of the compound of interest
Cx is Concentration of the compound
Ais is Area of the internal standard
Cis is Concentration of the internal standard

Once the response factor is established, analysis of an unknown mixture involves adding a precisely known quantity of the internal standard and conducting chromatography.

167
Q

What does the external standard method (chromatography quantitation) involve?

A

Automated sample injection systems and multiport injection valves in High-Performance Liquid Chromatography (HPLC) exhibit excellent reproducibility, enabling a sequence of injections with minimal variation in sample volume, typically less than 1%.

In the calibration process, a series of standard mixtures with precisely known concentrations of the analytes is analyzed, and their corresponding peak areas are recorded.

This data is then used to construct a calibration graph plotting the area against the concentration for each analyte. The purpose of this graph is to verify a linear response from the detector concerning the concentration of the analyte.
Once confirmed, this calibration graph serves as a reference tool for determining the amount of the analyte present in a mixture based on its detected peak area.

168
Q

What does the standard addition method (chromatography quantitation) involve?

A

This process involves adding a standard solution of a known analyte concentration to an unknown solution. Typically, multiple solutions are prepared, each containing the same volume of the unknown solution but varying amounts of the standard. For instance, using five 25 mL volumetric flasks, 10 mL of the unknown solution is added to each.
Then, different volumes of the standard solution (e.g., 0, 1, 2, 3, and 4 mL) are added to each flask. The mixtures are subsequently diluted to the mark and thoroughly mixed.

The principle behind this procedure lies in combining the unknown and standard solutions to create varying total concentrations of the analyte. The expectation is that the total concentration varies linearly with the added standard volume.
If the signal response from the detector shows linearity within this concentration range, it generates a plot resembling the one demonstrated. This plot aids in determining the concentration of the analyte in the unknown solution based on the known concentrations of the standard and the resulting signal response.

169
Q

How does flash chromatography differ from preparative chromatography?

A

Flash chromatography is a form of preparative chromatography.

This method differs from conventional techniques in two ways:

  1. It uses slightly smaller silica gel particles.
  2. Due to the restricted flow of solvent caused by the smaller gel particles, pressurized gas (nitrogen or air at approximately 10-15 psi) is used to drive the solvent through the column of stationary phase.

As a result, this approach enables rapid and high-resolution chromatography, often completing the process swiftly (“over in a flash”).

170
Q

How can a chromatography system be overloaded?

A
  1. By increasing sample conc whilst maintaining a constant injection volume (conc overload). Column efficiency has little effect on conc overloading.
  2. By increasing the injection volume whilst maintaining a constant sample conc (volume overload). Volume overloading is heavily influenced by stationary-phase particle size and column diameter.

A column is considered overloaded when retention factors obtained at low sample concentrations, change by more than 10% as the sample size is increased.

171
Q

Mixing checklist considerations:

A
  • What is the mixing duty?
  • What is the volume?
  • What is the success criterion?
  • Operation mode:
    – batch, semi-batch or continuous?
  • Operating conditions:
    – flow rates, temperature, pressure,…
  • Chemical reaction:
    – reaction rates
  • What phases are present?
    – Volume fraction?
    – Physical properties
  • How do physical properties change during run?
  • Sketch the vessel, including all major
    dimensions
  • What impellers are fitted?
    – What sizes?
    – Motor/gearbox ratings?
  • What vessel internals are present?
    – Baffles
    – Inlet and outlet pipes?
    – Dip pipes?
    – Surface additions?
    – Solids bed depth
    – Gas-liquid dispersion height
    – Heating/cooling coils
  • Solids-liquid mixing
    – Particle size, density, concentration
    – Solids distribution
    – Solids degradation
    – Agglomeration?
  • Liquid-liquid mixing
    – Interfacial tension
    – Volume fraction of liquids
    – Mass transfer with chemical reaction?
    – Phase inversion
  • Heat transfer
    – Liquid heat capacity
    – Thermal conductivity
    – Temperature of heating/cooling medium
    – Heat load and transfer area
172
Q

Mixing mechanical design checklist:

A
  • Power draw P must be known for sizing the motor and drive
    – Large power draw means large drive
  • Gearbox is rated to run over a range of N and torque Λ
    – Torque determines the gearbox, hence the capital cost
  • Gearboxes are more expensive than motors!
  • P and Λ determine the capital and running costs
    – If the impeller must start from rest in a bed of solids, the power and torque
    requirements for start-up can be extremely large, and depend on the characteristics of the solid
  • Power dissipated by the impeller is important for predicting process performance:
    – Mixing time, mass transfer coefficient and droplet sizes have been correlated to power
    – Power consumption calculated from torque measurements: P=2 π N Λ
  • Torque is measured in different ways at different scales
    – Small scale measurements
  • Mount vessel or motor on frictionless bearings and measure torque using a load cell or dynamometer
  • Use a commercial torque meter
  • Use a modified rheometer
    – At larger scales:
  • Strain gauges can be used.
  • Electrical current can be used.
    – Subject to many errors
    – Estimate of losses in the gearbox and bearings required
  • Beware of:
    – Vibration & resonance problems
173
Q

How is average slurry density calculated?

A

ρ.ave = (ms + ml)/V

Where:
ms - mass of solids
ml - mass of liquids
V - volume

If their densities and solid concentration is given,
ρ.ave = ρl + ((ρs - ρl)x)/((ρs/ρl)(1-x)+x)

174
Q

What is the Zwietering correlation?

What is the Zwietering correlation equation?

A

The Zwietering correlation refers to an equation used to estimate the mixing time required to achieve a certain level of homogeneity in a tank or vessel.

N.js = S * v^0.1 * dp^0.2 * X^0.13 * D^-0.85 * ((g*dρ)/ρl)

Where:
N.js - just suspension speed
S - vessel related constant
v - kinematic viscosity
dp - particle diameter
g - gravity
dρ - density difference between solid and liquid
ρl - liquid density
X - solid to liquid mass ratio
D - impeller diameter

N.homogeneous ≈ 1.25 * N.js

175
Q

What are the 5 “million dollar questions” in pharma drug development?

A

1) What are we making?
- Tablets (size, shape, coating…)?
- Capsules?
- Suspension?
- Aerosol?
- something else?

2) How much active ingredient and in what form?
- particle size distribution?
- crystalline vs. amorphous?
- pure API vs. salt vs. co-crystal vs. solvate?

3) What are the requirements on dissolution / release kinetics?
- immediate release vs. sustained release?
- pH dependent release? (mouth, stomach, intestine)
- t10, t50, t90 ?

4) What is the overall composition of the product (formulation)?

5) What is the process route (sequence of unit operations)?

176
Q

Main steps of tabletting:

A

Cyclic operation:
- Feed
- Pre-compact
- De-aerate
- Final compaction
- Eject

Need to use lubricants (e.g. magnesium
stearate) to avoid sticking to punch or die walls and reduce wear.

Problems:
capping, de-lamination

177
Q

Describe extrusion-spheronisation:

A

Principle of operation:
-Prepare a paste from API, excipients, binder, water
-Extrude paste through screen to form “noodles”
-Contact extrudates with a high-speed rotating disk
-Dry resulting spherical pellets

178
Q

Two plant layout approaches:

A

Vertical flow principle:
- processing stations on different levels
- gravity flow

Horizontal flow principle:
- processing stations on the same floor
- transport by IBC’s or pneumatic conveying system

179
Q

Regarding particle size-dependent solubility, how is the solubility of a particle with radius r calculated?

A

c(r) = C(∞)e^((2γVm)/(rR*T))

Where:
c(r) - solubility of particle with radius r
c∞ - solubility of macroscopic particle
γ - solid-liquid interfacial tension
Vm - molar volume of solid
R - molar gas constant
T - temperature

Large particles will grow at the expense of small ones (Ostwald ripening)

180
Q

What is the meaning of a critical radius of a particle?

A

The smallest crystal that can exist in a solution of given concentration.

Typically 4-10 nm for many solids.

181
Q

List key crystal growth regimes:

A
  1. Continuous growth (many growth sites on surface)
  2. Surface nucleation (several nuclei on surface)
  3. Spiral growth (single nucleus on surface)
182
Q

When carrying out mass balances on crystallisers, what do n and G represent?

A

G - growth rate (um / s)

n - number density (# /um /m3)

Plotting size (um) vs ln n gives a linear (-ve) plot (with a slope of -F/VG, with F/V = 1/t) and a y intercept of ln n0.

183
Q

What is Darcy’s law for filtration?

A

An empirical linear relationship between the macroscopic filtration velocity, and the macroscopic pressure gradient.

ΔP = (Rf + Rc)(µ/A)dV/dt
Where:
µ - viscosity
A - CSA for flow
Rf - filter resistance
Rc - cake resistance (hc/kc)
kc - cake permeability

184
Q

How is specific filter cake resistance, a, found?

A

a = A*Rc / ms

A - CSA for fluid flow
Rc - cake resistance
ms - mass of cake

185
Q

How is dry cake mass per filtrate volume, x, found?

A

x = ms / V

Where:
ms - mass of cake
V - filtrate volume

186
Q

What is the equation for general filtration?

A

dt/dV = µαxV/(A2∆P) + µRf/(A∆P)

Where:
V - filtrate volume
µ - fluid viscosity
α - specific cake resistance
x - dry cake mass per filtrate volume
Rf - filter resistance
A - CSA for fluid flow

SIMPLIFIED VERSION:
dV/dt = kA∆P/(µ*L)

dV/dt - rate of volume change over time (filtration rate).
k - permeability of the filter medium.
A - effective filtration area.
ΔP is the pressure drop across the filter.
μ is the viscosity of the fluid.
L is the thickness or length of the filter medium.

187
Q

What’s the solution for the general filtration equation for constant pressure, ∆P?

A

Equation:
dt/dV = µαxV/(A2∆P) + µRf/(A∆P)
∆P = const.

Integrate the filtration equation…
[µαx / (2A2∆P)] V^2 + [µRf/(A∆P)] V - t = 0

t/V vs. V is a straight line ⇒ useful for data analysis

mass balance ⇒ cake depth as function of filtrate V:
hc = xV/[Aρs(1-ε)]

x - dry cake mass per filtrate volume
hc - cake thickness
ε - void fraction of filter cake

Plotting V vs time shows an upwards curve that plateaus after enough time.

188
Q

How is moisture ratio, φ, of filter cake found?

A

moisture ratio: φ = mass of wet cake / mass of dry cake

φ = 1 + (ρL/ρS)* ε/(1-ε)

189
Q

What’s the solution for the general filtration equation for constant dV/dt?

A

Start with general equation:
dt/dV = µαxV/(A2∆P) + µRf/(A∆P)

Rearrange…
∆P = (µαx/A2)(dV/dt) V + (µRf/A) dV/dt

Plotting ∆P vs time for constant rate, ∆P increases linearly with time until ∆P.max is reached, then plateaus and remains constant.

190
Q

DELETE

A
191
Q

Describe how droplet morphology may vary with skin and non-skin formation:

A

If particles do not form a skin, there will be nucleation, and drying will likely result in porous particles.

Regarding skin formation, if a “strong” skin is formed, smooth particles are formed (hollowed centres).
If a “weak” skin is formed, the droplet can buckle, forming collapsed / non-spherical particles.
Weak skin can also lead to “puffing”, there the skin actually breaks, leading to collapsed shells.

192
Q

List key unit operations in pharmaceutical processing related to i) chemistry, ii) particle forming, and iii) particle processing

A

Chemistry:
- Reactions
- Separations
- Environmental (mass balances and solvent recovery)

Particle forming:
- Crystallisation
- Isolation
- Drying

Processing:
- Sieving
- De-lumping
- Micronisation
- Formulation

193
Q

‘What to watch out for’ when scaling up a pharmaceutical process:

A

Chemical Incompatibilities
* Solvent / reagent incompatibilities
* Solid/solid interactions
* Peroxide formation
– Ethers
– Aldehydes and ketones
– Secondary and tertiary alcohols

Self-Heating
* Most organic materials are unstable
* Rate of decomposition increases with temperature
* If heat losses > heat generated, temperature doesn’t increase and rate of decomposition remains slow
* When heat generated > heat losses, temperature begins to rise

194
Q

What’s the scale up rule and constant parameter for solid distribution?

A

Equal energy input (P/V = ε)

m = 0.67 so, for N*D^m = constant,

Constant parameter = N*D^(2/3)

195
Q

What’s the scale up rule and constant parameter for gas-liquid processes?

A

Equal mass transfer (kLa = ε)

m = 0.67 so, for N*D^m = constant,

Constant parameter = N*D^(2/3)

196
Q

What’s the scale up rule and constant parameter for solid suspensions?

A

Zwietering [ N.js = S * v^0.1 * dp^0.2 * X^0.13 * D^-0.85 * ((g*dρ)/ρl) ]

m = 0.85 so, for N*D^m = constant,

Constant parameter = N.js*D^(0.85)

197
Q

What’s the scale up rule and constant parameter for liquid blending?

A

Equal tip speed U

m = 1 so, for N*D^m = constant,

Constant parameter = N*D

198
Q

What’s the scale up rule and constant parameter for fast reactions?

A

Equal mixing time

Constant parameter = N

199
Q

What’s the scale up rule and constant parameter for heat transfer?

A

Equal Re

Constant parameter = ND^2

200
Q

How does impeller speed differ upon scale up?

A

It decreases (the amount it decreases to can be determined by the [6] different scale-up rules)

201
Q

What are the 2 ways crystal growth can be expressed?

A
  1. By mass deposition rate
    R = K(G)Δc = 1/Adm/dt = 3ψ(v)ρ*G/ψ(A)

Where ψ(v) and ψ(A) are volume and surface shape factors

  1. Linear growth rate
    G = dx/dt
201
Q

How can primary nucleation rate be calculated?

A

B = K(B)e^((-16 γ^3Vm^2)/(3R^3T^3σ^2)

Where:
σ - supersaturation
γ - interfacial tension between crystal and solution
Vm - molar volume of crystal
K(B) - function of agitation rate, equipment type, viscosity, and density

202
Q
A
203
Q

Describe reverse phase chromatography:

A

It uses alkyl chains covalently bonded to a solid silica support.
This creates a hydrophobic stationary phase, which has a stronger affinity for hydrophobic (or non-polar) compounds.
The use of a hydrophobic stationary phase can be considered the opposite, or “reverse”, of normal phase chromatography - hence the term “reversed phase chromatography”.
The non-polar stationary phase is used with a polar mobile phase to separate solutes based on their molecular polarity.

More polar compounds will interact less with the stationary phase and will be retained less than non-polar solutes.

204
Q

Explain the relevant underpinning scientific concepts of the retention factor, and derive the
mathematical expression:

A
  • The retention time, tr, is the time taken for a compound to travel through a column (from when it was injected to when it reaches the detector)
  • If no compound is retained, it will still take time to travel through the column.
  • The retention factor, k, is the time that a solute spends interacting with the stationary phase, found by correcting the retention time with the dead time, t0.

k = (tr-t0)/t0

205
Q

Describe relative retention time (RRT) and its calculation:

A

Relative retention time (RRT) reduces effects of variables that effect retention time, e.g. flow rate, temperature, and composition

RRT is an expression of a sample component’s retention time relative to the standard compound’s retention time.

A known compound is added to the sample matrix (an internal standard), and the relative retention times of the components is measured by:

RRT = Standard RT / Sample RT

Where:
- Sample RT is the retention time from injection of the sample of interest
- Standard RT is the RT measured from injection of the internal standard compound

206
Q

Describe the response factor and how it is calculated:

A

The response factor quantifies the relationship between the conc’ of a compound of interest and its response (typically measured as peak area or peak height) in the chromatogram.

The use on an internal standard procedure is recommended for accurate work, eliminating the need for accurate injections size a reference standard is included in each sample.
Using an internal standard, a response factor, RF, can be calculated to quantify the amounts of identified or known components.

RF = (Ax * Cis)/(Ais * Cx)

Ax is Area of the compound of interest
Cx is Concentration of the compound
Ais is Area of the internal standard
Cis is Concentration of the internal standard

207
Q

What’s the power number equation?

A

P = Po ρ N^3 D^5

Where:
Po - power number
Rho - fluid density
N - impeller rotational speed
D - impeller diameter

Po, or Newton number, Ne – Depends on
* Impeller type
* Impeller and vessel dimensions
* Properties of the phases present

208
Q

What does the aspect ratio of a vessel refer to?

A

The ratio of tank height (H) to tank width or diameter (T)

H : T

I.e. an aspect ratio of 1 means that H = 1

209
Q

How is the Penney diagram used?

A

The Penney diagram is a graphical representation for vessel scale-up purposes.

In the Penney diagram, the S^3 axis represents the scale-up factor cubed.
The scale-up factor is the ratio of the volume of the larger vessel to the volume of the smaller vessel.

The log P2/P1 axis represents the logarithm of the ratio of the power input per unit volume (P) in the larger vessel to the power input per unit volume in the smaller vessel.
This ratio helps in understanding the changes in power requirements as vessels are scaled up.

The Penney diagram allows engineers to determine whether the mixing conditions in the scaled-up vessel will be similar to those in the original vessel.
Generally, if the points representing different scale-up scenarios fall along the same curve on the Penney diagram, it suggests geometric similarity in mixing conditions between the vessels.

Here’s a summary of how the Penney diagram works:

Plot points on the Penney diagram for different scale-up scenarios, where each point represents a combination of scale-up factor and power input ratio.
Analyze the distribution of points on the diagram. If the points fall along the same curve, it indicates geometric similarity in mixing conditions.

210
Q

What are the 5 lines typically shown on the Penney diagram when considering scale up, in order of greatest to lowest gradient?

A
  1. Constant mixing time (+ve gradient)
  2. Constant P/V (y = 1, horizontal line)
  3. Just suspension / Zwietering (-ve gradient)
  4. Constant tip speed (more -ve gradient)
  5. Constant heat transfer / Re (steep -ve gradient)

All line start from (0,1)

211
Q

What are the main steps involved in Direct Compression and Dry Granulation tablet manufacturing techniques?

A

Direct Compression Route: API + Filler + Lubricant > Size reduction > Blending > Tabletting > Coating (+ polymer solution) = Finished tablets

Dry Granulation Route: API + Filler > Blending > Roller compaction > Milling and Sieving > Blending (+ lubricant) > Tabletting > Coating (+ polymer solution) = Finished tablets

Unit operations:
Blending – forms homogeneous blend of API and excipients
Roller compaction – prevents segregation and forms plastically deformable particles suitable for tabletting. Improves bulk density and powder flowability
Sieving – removes oversized particles after dry granulation
Tabletting – forms actual tablets from the granules or powder blend
Coating – applied for taste masking due to bitter taste and unattractive appearance of the API

212
Q

What are the main steps involved in wet granulation tablet production?

A

API + Filler > Mix/Dissolve/Suspend > Dry > Milling and Sieving > Blending (+ lubricant) > Tabletting > Coating (+ polymer solution) = Finished tablets

213
Q

What are the main unit operations in tablet formation?

A

Blending – forms homogeneous blend of API and excipients

Roller compaction – prevents segregation and forms plastically deformable particles suitable for tabletting. Improves bulk density and powder flowability

Sieving – removes oversized particles after dry granulation

Tabletting – forms actual tablets from the granules or powder blend

Coating – applied for taste masking due to bitter taste and unattractive appearance of the API

214
Q

List tablet excipients and their functions:

A

Binder – improves tablet compressibility and mechanical properties
Filler – improves blend homogeneity and compatibility
Disintegrant – improve tablet break-up during dissolution
Lubricant – helps avoid material sticking to the tablet punch
Desiccant – binds to moisture in the packaging to avoid API degradation
Coating material – taste masking and improved appearance

215
Q

List suitable impeller designs for axial and radial flow:

A

Axial:
- 45° pitched blade
- Hydrofoil
- Propeller

Radial:
- Rushton turbine
- Smith turbine
- Disc turbine
- 45° pitched blade
- Anchor

216
Q

What are the key aims of the FDA?

A
  • Promote new technology
  • Introduce modern quality management techniques
  • Risk-based approaches to R&D and inspection
  • Promote science-based policies
  • Improve FDA drug quality regulatory programs

FDA wants to move from a product centred regulatory program to a systems based regulatory programme, thereby easing the regulatory burden for both FDA and industry.

  • Regulators approve investigative new drug applications (IND) as well as new drug applications (NDA).
  • INDs feeds a company’s research pipeline and ensures business sustainability
  • NDAs create revenue to fund R&D and provide return on investment for shareholders
217
Q

How is Reynolds number calculated for pharma processes with impellers?

A

Re = ρND^2 / μ

Where:
μ is viscosity (Pa s)
N is rotational speed (rps)
D is diameter (m)
ρ is density (kg/m3)

218
Q

Chromatography is an essential analytical method during pharmaceutical process development and there are different methods to obtain quantitative information from chromatograms.
In your opinion, which one is the most popular one in a synthetic chemistry laboratory during API route selection (early development) and which one is preferred by chemical engineers for kinetic studies (late development)?

Give reasons for your decision:

A

There are 4 main methods for identifying quantitative information:
- Normalizing peak areas
- Internal standards
- External standards
- Standard addition

  • Normalising peak areas is the most popular with synthetic chemists.
  • The area of an individual peak is calculated as a percentage of the total areas recorded for all peaks.
  • It is simple and quick
  • During route selection, there will be lots of samples, and the basic information of peak area normalisation is sufficient to make decisions on the route as it relies on major effects being seen.
  • However, it does not reliably provide information on absolute concentrations, hence it is useless for kinetic studies in late development.
  • For kinetic studies, internal standards, external standards, and standard addition can be used.
  • To minimise sample handling and the associated inaccuracies due to volume additions and variations in injections, the internal standard method would be preferred for kinetic work.
  • The internal standard method involves using a reference standard included in each sample.
219
Q

Clearly articulating a design space in a new drug application is key to receiving the regulatory authority’s approval on the manufacturing process, and an image says more than a thousand words.
How would you graphically demonstrate that your process performs reliably to expectations?

A

Technically, any diagram that compares experimental and predicted data can be used as evidence that the plant’s control strategies are working.

However, the best way to present this info is through parallel coordinate plots, allowing multidimensional data to be visualised in 2D

In the parallel coordinates plot, equally spaced parallel axes are drawn for each variable (CQA, QCPP, etc.)

Then a given row of data (values for a single batch) are represented by drawing a line that connects the values of that row on each corresponding axis

Proven acceptable ranges (PARs) can be displayed for single or multiple unit operations

As long as each batch (or overall process) always remains within the visual boundaries, there is evidence that the control strategy is working.

220
Q

In a secondary manufacturing plant, a quality control audit has revealed that some tablets do not have sufficient mechanical stability (i.e. break easily in half) and that the API is unevenly distributed in the tablet (i.e. each half contains a different amount of API).
After receiving the API from the primary process plant, it gets fed into a micronizer before the tablets are manufactured through a straightforward direct compression route.

Before being packaged, the tablets get coated.

a) Identify possible root causes of the problem in each unit operation and what should be investigated at each stage.

b) Where in the overall process do you think is the most likely cause for the fault?

A

a) Direct compression route has the following unit operations which could bring about an error:

  1. Milling – API could be under or over reduced in particle size at the mill stage. Check PSD before and after microniser.
  2. Blending – API gets blended with filler, binder, and lubricant. Either the blender does not mix for long enough or its efficiency is decreased somehow. A faulty solid addition system can contribute to error as the tablets have insufficient mechanical stability. Variance in composition should be checked here.
  3. Compacting – in the tablet press itself, 2 sources of error could occur - an insufficient compression force or insufficient ejection which leads to impurities and changes to the composition. Operation parameters and cleanliness of the press should be investigated here.
  4. Coating – does not usually impact mechanical stability unless pellets get too wet, leading to recrystallisation or dissolution. Drying rates should be checked.

b) The most likely source of error is in the blending stage (insufficient solid mixing) and the tabletting stage (solid cohesion is insufficient). A final possibility is that the API itself has changed e.g. into a different polymorph with difficult physical properties.

221
Q

List some difficulties of batch processing:

A

Longer reaction times
By-product formation
Frothing
Unexpected rheological changes
Slow filtrations
Slow phase separations
Lower yields
More impurities

222
Q

What is the smallest lab scale for which you can accurately investigate (e.g. mixing behaviour)?

A

2L

223
Q

How does equal power input P/V relate to degree of agitation?

A

0.005 - gentle
0.1 - mild
0.3 - moderate
1.0 - high
2.0 - intense

P/V values in kW/m3

224
Q

What is the baseline resolution for chromatography?

A

1.5

Baseline resolution usually occurs at resolution number of 1.50; however, there is no visible baseline between the two peaks.

Numbers greater than 1.50 indicate there is baseline between the peaks and numbers less than 1.50 indicate there is some degree of co-elution.

225
Q

How is selectivity of a chromatography device found?

A

a = k2 / k1

Where k1 and k2 are the retention factors for species 1 and 2.