2202 CSV Flashcards

(325 cards)

1
Q

Explain glycerin-gelatin mixtures as suppository bases

A
  • Gel formed from gelatin, glycerin and water (14 %w/w gelatin, 70 %w/w glycerin ,16 %w/w water)
  • Gelatin content can be increased for a stiffer consistency
  • Good for water-soluble drugs
  • Glycerin is a laxative
  • Gelatin type A = cationic and gelatin type B = anionic, therefore care with charged drugs
  • Water present, therefore preservative required
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Explain polyethylene glycol as a suppository base

A
  • Low molecular weight (MW) PEGs (200 to 600) are liquid
  • High MW PEGs (>1500) are solid
  • Can mix MWs to give desired properties
  • PEG acts as a cosolvent
  • Will absorb water from the rectal mucosa, causing
    stinging. Therefore, include water in the formulation
    (up to 20 %w/w)
  • Water present, therefore preservative required
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Explain synthetic triglycerides as suppository bases

A

Hydrogenated vegetable oils, e.g. Witepsol family
- range with different melting points and viscosities
- select the appropriate one for the drug used
- H15 for insoluble drugs, E75 for liquids/soluble drugs

  • Safe, non-toxic, non-irritating
  • Easy to work with, can be reheated
  • Little batch-to-batch variability
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

outline the ideal properties of a suppository base

A
  • Melts at or just below body temperature (but solid at ambient temperatures) or dissolves in rectal fluid
  • Non-toxic and non-irritating
  • Therapeutically inert
  • Compatible with all drugs, i.e. chemically inert
  • Physically and chemically stable
  • Easy to work with
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

what are the usual sizes and age ranges for the suppositories?

A
  • 1 g (babies and small children)
  • 2 g (older children and adults)
  • 4 g (adults)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What conditions are in the rectum/

A

pH is essentially neutral, low levels of fluid, some mucus

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

what is rectal foam?

A

dispersion of gas in a liquid, stabilised by surfactants

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

what two ways does the suppository ‘breakdown’ in the rectum?

A
  • Suppository base melts at or just below body temp, e.g. Witepsol H15, theobroma oil
    OR
  • Suppository base dissolves in rectal fluid, e.g. glycerin- gelatin, PEG
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

how does breath holding influence ventilation?

A

• Enhances deposition by facilitating gravitational sedimentation
• Breath-holding for at least 4 sec important
• Breath-holding is important for smaller particle sizes as they have an increased chance of being exhaled because they can remain airborne for a considerable time

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

how does particle size effect aerosol deposition?

A
  • smaller particles achieve greater total lung deposition, farther distal proposition and more peripheral deposition
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

how does the size of the inspired volume effect ventilation?

A

• Amount of aerosol that enters the lung
• Depth of penetration of the aerosol into the lung

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

how does the timing of aerosol administration effect ventilation?

A

• Delivery of the aerosol early in inspiration puts the aerosol at the “front end” of the breath for deeper penetration
• Delivery of the aerosol later in inspiration puts the aerosol at the “back end” for delivery to later-filling lung units

  • just start to breath and then hit the button and continue to breath in.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

what 3 factors effect aerosol deposition?

A
  1. Physical properties of the aerosol particles
  2. Ventilation factors
  3. Regional airway anatomy / lung mechanics
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

what are some advantages of aerosol delivery?

A

• Alternative to injection
• Non-invasive
• Relatively convenient
• Direct delivery of drugs for respiratory disease
• Rapid onset of action
• Lung provides a large surface area and a readily accessible blood supply
• Delivery route to systemic circulation

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

what are some ventilation factors?

A

• Particle velocity
• Inspiratory flow
• Inspired volume
• Breath-hold duration
• Timing of aerosol delivery during inspiration

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

what are the mechanisms of deposition?

A

• Inertial impaction
• Gravitational sedimentation
• Brownian diffusion
• Electrostatic precipitation

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

what is an aerosol/

A

Any system of solid or liquid particles of sufficiently small diameter to maintain some stability as a suspension in air

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

what is brownian diffusion

A

• Small particles are displaced by the random bombardment of gas molecules and collide with the airway walls
• These very small aerosols deposit in the distal lung
• This is important for particles <0.5 μm

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

what is brownian precipitation?

A
  • The charge on an aerosol can enhance the deposition of very small aerosols that have a high mobility
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

what is gravitational sedimentation?

A

• Occurs for smaller particles that are able to follow the airstream and penetrate the more peripheral bronchioles and alveoli (airstream flows slower here)
• Particles <2 μm deposit in the lower airways by gravitational sedimentation

this is why we ask patients to hold their breath for as long as comfortable after using their inhaler

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

what is inertial impaction?

A

• Refers to particles that tend to continue in their present trajectory rather than follow the curvature of the airways i.e. impact the airway wall
• Mechanism dominant in the oropharynx and major bronchi for rapidly inhaled particles >5 μm
• To impact a surface, the particles must have enough inertial momentum to overcome the drag force of the gas flow on the particles

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

what is inspiratory flow and how does it effect ventilation?

A

• Typical inspiratory flows during quiet tidal breathing ~ 0.25-0.5 L/s
• Faster inspiratory flows increase oropharyngeal and upper airway deposition by inertial impaction
• Slower inspiratory flows minimise oropharyngeal and upper airway deposition and enhance distal delivery through gravitational sedimentation

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

what is particle velocity and how does it effect ventilation?

A

• Determined by the aerosol generator (Lecture 2)
• The basic function of an aerosol generator is to generate a drug-containing aerosol cloud that contains the highest possible fraction of particles in the desired size range
• Pressurised metered-dose inhaler particle velocities range from 10-100 m/s

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

what is the definition of aerodynamic diameter?

A

The diameter of a unit density sphere with the same settling
velocity as the particle in question.

(Basically, this means that we take the physical measured value and determine the size of the sphere that could produce the data)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
how do dry powder inhalers work? Give some advantages and disadvantages.
- fine particles +/- an inert carrier eg lactose Advantages - less patient coordination needed, breath holding not needed and no propellant required Disadvantages - requires moderate to high inspiratory flow, some units are single dose, difficult to deliver high doses
26
how do nebulisers work?
- Compressed gas or an ultrasonic device is used to convert a liquid into a fine mist - Drug is breathed in through a facemask or mouthpiece via a connecting tube - Two types: jet nebuliser + ultrasonic nebuliser
27
how do the soft mist inhalers work?
• Multi-dose, propellant-free, liquid inhaler • Higher fraction of fine particles than pMDIs and DPIs • Aerosol spray exits slowly and for a longer duration than pMDIs • Higher lung / lower oropharyngeal deposition • Lower doses without lowering efficacy
28
outline how the pressurised metered dose inhaler works and som advantages and disadvantages.
• Drug dissolved in liquefied propellant-based formulations • The propellant is a gaseous compound which pushes the product out of the container and produces a spray Advantages = convenient, difficult to contaminate, portable Disadvantages = patient coordination is vital, difficult to deliver high doses
29
what are the 4 mechanisms for clearance e of deposited aerosols?
1. Absorption into the bloodstream 2. Local inactivation = accomplished through macrophage injection 3. Lymphatic clearance = absorbed into lung tissue + cleared through lymphatic drainage 4. mucociliary transport = occurs if the drug is deposited on the airway surface and then swallowed
30
what are the 4 types of aerosol generators?
1. Pressurised metered-dose inhalers (pMDIs) • With holding chamber (spacer) 2. Nebulisers • Jet • Ultrasonic 3. Dry powder inhalers (DPIs) 4. Soft mist inhalers
31
why is HFA now used instead of CFC as a propellant?
- they don’t contribute to climate change - They have good safety - they are chemically stable; non-inflammable dose monitoring required when patients where switching from CFC to HFA as they had different lung distribution (HFA particles where smaller therefore could get deeper into the lungs) they where them matched mcg to mcg to CFC by changing (lowering) the amount of drug in the chamber
32
Outline reasons for solubilisation of drugs.
- Improved efficacy, e.g. increased bioavailability from aqueous solutions - Easier handling, e.g. easier dose measurement, reduced viscosity of aqueous systems - Reduced side effects, e.g. intramuscular parenteral suspensions are painful, non-precipitation of drugs from intravenous parenteral formulations - Marketing (aesthetics)
33
outline solubilisation by surfactant systems.
- Formulation of poorly water-soluble drugs in aqueous environments may be enhanced by the use of surface-active agent(s) at concentrations above their critical micelle concentration (cmc). - The surfactant acts as a ‘bridge’ between aqueous and non- aqueous environments and the drug may be solubilised by being held within the micelle or the outer, more hydrophilic long-chain structures (palisade layer). - Solubilising surfactants have a greater hydrophilic character (HLB>16) compared, e.g. to emulsifying surfactants. Examples include polyoxyethylene sorbitan mono-oleate (Tween 80) and monolaurate (Tween 20), polyoxyethylene lauryl ether (Brij 35).
34
outlines solutes for solubilisation.
1. polar solutes (drugs) 2. Non-polar solutes (drugs) 3. semi-polar solutes (drugs)
35
what are some techniques of altering drug solubilisation?
- surfactants - co solvents - complexation - modification of solid state - chemical modification - synthesis of enhanced solubility derivatives
36
what would the case of DD + WW > 2DW mean?
- that there will be less tham complete mixing and the drug will have a finite solubility in water. - the greater the difference the lower the drug will be soluble in aqueous solvent
37
what are advantages of a suspension?
• Drug insoluble in the delivery vehicle • Mask bitter taste of the drug • Peadiatric medicines • Increase drug stability • Patient unable to swallow solid dosage form
38
what are disadvantages of a suspension?
• Physical stability – sedimentation and compaction • Bulky formulation – in comparison solid dosage forms • 500 ml & 1000 ml bottles not uncommon • Uniform and accurate doses cannot be achieved unless formulation is well shaken just before use
39
what are the types of suspensions?
1. colloidal - 1 nm to 1 μm • Tyndall effect – light scattering 2. coarse - 1 to 100 μm • More liable to settling ou
40
what do vehicle and suspending agents do?
Aqueous solutions that increase the viscosity of the formulation Used in deflocculated systems
41
what do wetting agents do?
used in flocculated systems Some insoluble solids easily wetted by water and will disperse readily throughout the aq. phase with minimal agitation
42
what is sedimentation?
• Sedimentation of particles or floccules as a result of gravitational forces in the liquid • Flocculated suspensions – flocs form and increase sedimentation rate • Form loose porous sediment • Deflocculated suspension – individual particles begin to sediment • Form dense cakes
43
What is the definition of a suspension?
Heterogeneous mixture in which insoluble solid particles are dispersed evenly in a liquid medium
44
list drying methods.
1. convection drying 2. conduction drying 3. Spray drying 4. Freeze drying
45
outline conductive drying.
- Wet solid on direct contact with hot surface, hence heat transfer occurs via conduction - Most important example - vacuum oven
46
outline convection drying
• Drying takes place via latent heat of evaporation provided via a hot air stream • May be fixed bed - air passes over static powder mass •tray drying • May also be dynamic - powder is in motion •fluid bed drying
47
outline fluidized bed drier.
• Hot air stream through powder bed gives buoyancy to particles • Particles form a fluidised suspension in air • Water loss takes place from individual particles
48
outline freeze drying
• Method used to produce dried product (from aqueous solutions or suspensions in vials) • Particularly important because it involves the use of low temperatures • Results in porous solid • Widely used for the preparation of proteins
49
outline spray drying.
• Method used to produce dried product from aqueous solutions or suspensions • Solution atomised (liquid converted to small droplets) and sprayed into stream of hot air • Results in free -flowing powder sample
50
what are the different types of water in a solid sample?
• Water of crystallisation (e.g. hydrates) • ‘Free’ water - water essentially present as a liquid • ‘Bound’ water - hydrogen bonded to the substrate
51
what is caking?
• Inappropriate choice of drying conditions can lead to rapid water loss from surface of material which forms a hard dry crust (caking) • This caked material then prevents further water loss from deeper in the bed while the now dry crust becomes hot
52
what is constant rate period?
- Sample loses water from surface of bed and saturates air immediately above surface . - Water replaced immediately, hence water is effectively being lost from a free liquid surface - Rate will therefore be constant, as it is limited by rate of vapour removal from above bed - Controlled largely by rate at which water vapour can be removed
53
what is drying? Why is it important?
removal of some or all of a solvent from a system - Pharmaceutical importance: • chemical stability • processing and handling • toxicity • dosing
54
what is first falling rate period?
• Rate of vaporisation not sufficient to • saturate air above surface • Drying rate determined by rate of transfer to surface. • As drying proceeds, this becomes increasingly difficult hence rate decreases with time • Rate of transfer determined by diffusion, suction potential of porous materials and capillary force
55
what is second falling rate?
• In first falling rate period, water is still being lost from surface. • In second falling rate period, water loss is from within bed itself, i.e. no more free water • As solid becomes dryer thermal conductivity also decreases, • may need to increase bed temperature (an issue for thermolabile materials) • In practice, first and second falling rate periods seen in continuity
56
outline the mechanism of the transcellular route by passive diffusion.
• High concentration of drug on apical side of cell • Low concentration inside the cell • Molecule diffuses into cell and out the other side into intercellular area and the blood - many lipophilic drugs and neutral molecules
57
what 4 ways can drugs be absorbed from the GI tract?
• Transcellular route by passive diffusion • Transcellular route via active transport • Transcellular route via facilitated diffusion • Paracellular route (tight junctions)
58
what are the functions of the large intestine?
- limited absorption for drugs - absorbs water - absorbs lipids - gut microbiome will metabolise many nutrients and drugs
59
what does the small intestine do?
- absorbs nutrients, salts + drug molecules.
60
what generally occurs in the stomach/
- mechanical digestion of food = peristalsis - mixes the food with gastric juices = chyme - this facilitates the absorption of nutrients
61
what is acid, pepsin and mucus in the stomach produced by? And what do they do?
- parietal cells (kills bacteria / aids breakdown) - peptic cells (breaks down protein) - mucosal cells (protects the stomach from auto digestion)
62
what is enterohepatic
• Bile salts are produced in the liver stored in the gallbladder • They are released in the duodenum in the presence of food, where they act as surfactants and emulsifiers • Most of the quantity of bile salts is reabsorbed (mainly in the ileum) and stored back in the gallbladder to be reused
63
what is the definition of biopharmaceutics?
Study of the physical and chemical properties of drugs, their route of administration, and how these affect the rate and extent of absorption
64
what is the result of first pass metabolism?
• Reduction in the amount of drug enteringvthe systemic circulation • Oral dose needs to be higher when a drugvundergoes extensive first-pass metabolism • This effect limits bioavailabilit
65
where do all the veins leaving the GI tract which carry blood rich in nutrients and drug drain to?
hepatic portal vein, taking drug compounds to the liver where they undergo 1st pass metabolism
66
are acidic drugs ionised at low or high pH
High pH
67
outline the mechanism for dissolution controlled release system.
• The drug is covered with a slowly dissolving coating • The liquid surrounding the unit dissolves the coating • Rate-limiting dissolution step • Solid drug is exposed to the liquid and dissolves
68
outline the mechanism of diffusion controlled release systems. (matrix system)
• Drug is dispersed as solid particles within a matrix formed of a water-insoluble polymer (e.g., poly(vinyl chloride) or in a matrix forming a gel in contact with water • Release from a matrix system involves: 1. Drug particles located at the surface will be dissolved and released rapidly 2. Drug particles at increasing distances from the surface will be dissolved and released by diffusion in liquid- filled pores of the matrix or in the gel to the exterior of the release unit
69
outline the mechanism of diffusion controlled release systems. (Resivoir system)
Release of drug from a reservoir system involves: - Partition of the drug dissolved inside the release unit to the solid membrane - Transport by diffusion of the drug within the membrane - Drug will partition to the solution surrounding the release unit
70
outline the paracellular route by tight junctions.
Absorption through the gap between neighbouring cells – tight junctio
71
outline transcellular route by active transport.
- Molecule “piggybacks” into the cell using a system designed for natural substrates e.g. amino acids, vitamins - Molecules are moving against a concentration gradient - Therefore this process requires energy
72
outline transcellular route by facilitated diffusion.
• A form of passive transport • No energy is required • Uses carrier proteins • Requires a concentration gradient • Structurally selective • Competition of different drugs for binding sites • Saturation on apical side if not enough carrier
73
outline the 5 volumes of the BP.
Vol 1/2 =0.5 monnographs for medicinal and pharmaceutical substances. Vol 3 = monographs for formulated products Vol 4 = infrared reference spectra Vol 5 = veterinary
74
What is a pharmacopoeia.
Provide publicly available and legally enforceable quality control standards for common drugs
75
what is the difference between European and BP pharmacopeia?
EP has no monographs on individual formulated products, whereas the BP does!
76
what is the difference between the European pharmacopeia and the British pharmacopeia?
European pharmacopeia doesn’t contain any formulated drug monographs and is updated every few years, whereas the British Pharmacopoeia contains formulated drug monographs and is updated every year
77
outline factors that effect drug absorption.
• Stratum corneum barrier integrity compromised by disease. • Raised skin temperature (increased rate of drug diffusion). • Occlusion of skin (by formulation or applied dressing) - increases water content of stratum corneum by reducing transepidermal water loss, opening out tightly packed structure and decreasing resistance to diffusing drug. • Chemical manipulation of barrier (penetration enhancement). • Maximising drug concentration in solution in applied surface film (formulation)
78
outline the way a topical drud may act on the barrier stratum corneum.
- increasing the barrier (suncreams / barrier creams) - reducing the barrier efficiently (transdermal delivery) - treating a barrier damaged by skin disease or infection (antibiotics, steriods) - restoring the physical characteristics of a barier adversely affected by the environment (emollient / moisturuser)
79
what are the 5 layers of the epidermis? From the deepest
1. stratum germinativum = resting upon the basal lamina. Cells undergo mitosis - replacement of shed epidermis 2. Stratum spinosum = as prickles or spines extending from the surface of the cell. No mitosis but metabolically active. Spines link via intercellular bridges - improved structural rigidity and resistance to abrasion 3. Stratum granulosum = granular cells with granules of keratatohyalin, a precursor of keratin 4. Stratum lucidum = not always present/observed. Cells are elongated, translucent, lacking cytoplasmic organelles. 4.
80
what is the epidermis of the skin?
- outermost layer of the skin (in contact to environment) - Stratified epithelium, consisting of five distinct layers.
81
what is transdermal vs what is topical
when you apply a product to the skin with the intent for it to be systemically absorbed, for a whole body delivery. Whereas, topical is application to the skin with the effect of a local outcome, ie no systemic absorption = local effect
82
whats the surface area of the skin of an adult and how much blood does it receive?
Approximately 2m2 and 1/3 the blood
83
how do we classify pharmaceutical glass)
Type 1 (high resistance, least reactive, high cost) to type 4 (low resistance, most reactive lower cost_
84
Outline compliance and adherence.
1. compliance aid - blister packs, help patients remember to take their medication 2. Specialised packing for specific populations - eg arthritis friendly caps for easy opening 3. Portability - slim packing to fit in pocket 4. Dosage control - eg metred dose inhaler / insulin pens
85
outline packaging and information.
- clear labelling is important for patients - key information needed eg dosing and warnings - marketing and brand recognition
86
outline preservation and contamination.
- preservation against microbiological contamination (sterile packaging + air tight seals) - particularly important in injection (IV)
87
outline protection of pharmaceutical
- temperature control (increased temp increases rate of reactions) - moisture control (hydrolysis + supports growth of microorganisms + dilution of product) - light (photodegradation - especially UV light) - atmospheric gases (mainly carbon dioxide + oxygen (oxidation reactions))
88
what application does glass have?
primary packaging - vials, ampoules, bottles, jars - widely used for liquids, semi solids and solids
89
what are advantages and disadvantages of of glass?
Advantage: - easy to mould - cheap - easy to sterilise - impermeable to gas and moisture disadvantages: - fragile (easily broken) - heavy (transport costs)
90
what are the types of glass used?
1. soda glass (mostly silica - 72%) - reduced temperature needed to produce - improved mechanical strength and chemical durability - adds some amount of alkali to the water solutions (disadvantage) 2. Amber glass - similar to soda glass but iron oxide added - this gives colour which provides block to UV light absorption 3. Borosilicate - overcomes defects of soda glass by decreasing alkali - boric acid is very temperature resistant - very expensive to make (disadvantage)
91
what is hydrolytic resistance
Resistance to the release of soluble mineral substances into water under prescribed conditions of contact
92
what is pharmaceautical packaging
collection of different components which surround the pharmaceutical product from the time of its production to its use.
93
what is primary packaging?
- the packaging materials that are directly in contact with the product eg blister packs / strip foil packaging - it should not interact physically or chemically with the product
94
what is secondary packaging?
- pack component with no physical contact - usually made by paper or card - useful to contain information and labelling
95
what is tertiary packaging?
- further packaging that surrounds the secondary packaging - useful for logistics and supply chain management
96
what is the purpose of pharmaceutical packaging?
1. Protection and containment 2. Preservation 3. Presentation and information 4. Compliance
97
who regulates the pharmaceutical packaging?
MHRA, underpinned by the human medicines regulations 2012
98
Explain the general properties of thermoplastics
• Can withstand repeated thermal cycles to soften/melt/mould • Formed by techniques including injection moulding, blow moulding, extrusion, lamination • Usually resistant to breakage • Cheap to produce and therefore used more often than thermosets
99
outline aluminium.
• Foil for blister packs • Excellent barrier properties and can totally exclude moisture, oxygen/other gases, microorganisms, and light • This can help to maintain degradable products in peak condition for long periods • The manufacturing processes mean that the foil is sterile when formed • Other advantages: • Can be printed on • Flexibl
100
outline an example of extractable / leachables.
Leaching of DEHP from PVC bags (plasticising agent) - Plasticised PVC is used for IV and blood bags and infusion tubing, enteral and parenteral nutrition feeding bags, and tubing used in cardiopulmonary bypass devices - DEHP has been detected in liquids stored in plasticised PVC - DEHP has shown a wide range of toxic effects on the male neonate reproductive system development, as well as on lungs, heart and kidneys, and it has been classified as possibly carcinogenic by The International Agency for Research on Cancer
101
Outline plasticised PVC (thermoplastics)
- Plasticised PVC: The addition of a plasticise Disadvantages • Lowers strength • Lowers melting point • Decreases barrier properties • Leaching of DEHP associated with health concerns • Not easily recyclable Uses • Widely used for tubing, infusion bags and other similar medical devices
102
outline Polyethylene (thermoplastic) outline what increased / decreased density PE is associated with
- properties are relates to high and low density forms - this is the most common and economical packaging plastic 1. Increased density: • Greater rigidity • Higher distortion and melting temperatures • Lower gas and vapour permeability • Increased tendency to crack 2. Low density • LDPE is used for flexible packs e.g. bags, dropper bottles
103
Outline polypropylene (thermoplastic)
• Similar to PE, except every fourth H is replaced by a methyl (CH3) group • Suitable for rigid packs • Good resistance to most chemicals, except organic solvents • Similar barrier properties to HDPE • Better resistance to higher temperatures than HDPE, therefore more suitable for steam sterilisation • More brittle at lower temperatures than PE
104
outline problems with plastics and gas permeability.
• Gas molecules can diffuse through plastics • Different plastics have different gas permeabilities, with lower density plastics showing greater gas permeability • The addition of fillers (inert clays or fibres) can reduce permeability by lengthening the route of diffusion
105
outline rubber
• Used to produce air-tight closures for bottles, cap liners, and bulbs for dropper assemblies • Used in multi-dose vials and disposable syringes • Natural rubber, and neoprene and butyl/nitrile rubber (synthetic rubbers) are most common • Problems have been observed when rubber is in contact with parenteral solutions
106
outline some advantages and disadvantages of plastics in pharmaceutical packaging.
often where glass has disadvantages, plastics have the opposite advantages 1. Advantages • Good mechanical properties, even in thin-walled containers • Less brittle than glass • Relatively lightweight = reduced transport costs • Readily mouldable into various geometries • Suitable for containers and closures 2. Disadvantages • Majority will not withstand significant heat without distortion • Permeable to water vapour and atmospheric gases • May interact with certain chemicals in a manner that alters material properties • May absorb substances from solution • May release substances into solution • e.g. unpolymerised monomers or residues used in their processing
107
outline the mechanism of drug sorption.
• Rapid adsorption of the API onto the plastic surface, followed by partitioning into the plastic • Adsorption is an extremely rapid process whereas the rate of absorption is thought to be much slower • Rate determining step is therefore the diffusion of API into the plastic matrix • The loss of API from solutions stored in infusion bags is driven principally by the ability of the drug to diffuse into the plastic • Net effect: Loss of drug into packaging = loss of potency of product - drug adsorption onto plastic surface = fast - diffusion across plastic = slower (rate limiting step) = Lower concentration of API in solution therefore suboptimal dose
108
outline thermosetting plastics. Giving an example of this.
- they have a single processing window, and cannot be reshaped after curing - they are harder and tougher than thermoplastics - mostly used for rigid screw caps as cost limits their use example is plastic used for plug sockets
109
outline unplasticised Polyvinyl chloride (PVC) (thermoplastic)
• Clear/colourless • Good chemical resistance • Good barrier to moisture/gases • Low impact resistance • Use is diverse: bottles and blister packs
110
What are 5 factors affect drug sorption?
1. Surface area of the material available for absorption 2. Initial concentration of the API in solution 3. Diffusivity of the API into the PVC plastic 4. Partition coefficient (P) of API between the aqueous solution and the plastic material 5. The pH of the solution, which influences API ionisation
111
what are the two categories of plastics?
- thermosetting - thermoplastic
112
what does drug sorption mean?
• Drug can be adsorbed onto or absorbed into the packaging material • This can compromise its barrier properties and/or reduce product potency
113
what does extractable mean?
• Compounds that can be extracted from a packaging material under extreme conditions • e.g. exposure to solvent or elevated temperature
114
What does leachables mean?
• Compounds that can leach passively into the drug product formulation from the packaging material as a result of contact under normal conditions
115
what are the two typed of hard capsule closure?
Coni-Snap® • Tapered rim to aid closure • Air vents allow air to escape • Six dimples to provide pre-lock • Closely-matched locking rings Dbcaps® • Larger diameter, shorter length • The cap covers most of the body; impossible to hold the body and open without crushing it • Provides increased security of the contents
116
what is bloom strength?
measurement of gel rigidity (used to characterise the grade of gelatin) Defined as: The load/weight in grams required to push a plunger with a set bottom diameter (12.7 mm) a set distance (4 mm) into a 6.67% w/w gelatin gel that is prepared in water and allowed to mature at 10 °C for 16-18 h” high bloom strength = harder material and more viscous in solution
117
what is the difference between dependent and independent capsule filling methods?
1. dependent - • e.g. the auger system - Dosing systems that use the capsule body directly to measure the powder weight 2. Independent - e.g. the dosator system - Powder is measured independently of the body - Weight uniformity is not dependent on filling the body completely - Capsules can be part filled
118
what is the difference between gelatin band sealing and hydroalcoholic solvent seal? (Hard capsule sealing methods
1. Gelatin band sealing • A dilute solution of gelatin and colour pigment is applied to the centre of the capsule to hermetically seal it; this prevents leakage, contains odour, and reduces oxygen permeation 2. Hydroalcoholic solvent seal • 50:50 water/ethanol mixture is sprayed onto the joint between the cap and the body; excess is removed by suction • Capillary action draws the fluid into the joint and the presence of water softens the gelatin • Gentle heating (45°C) fuses the two layers fuse together, forming a sea
119
what is the difference between hard capsules and soft capsules?
1. hard capsules - two piece system consisting of a cap and body - locking device - most common 2. Soft capsules - one piece system eg docusate
120
why are capsules used since they are more expensive and harder to formulate than tablets?
- some tablets are very difficult to make as they can change polymorphic form after being compressed. - patient preference (easier to swallow) - suitable for drugs that need additional protection - combination therapies and biphasic release - masks taste and odour
121
why do we use gelatin to make the shell of capsules?
• Non-toxic & widely used in foodstuffs • Readily soluble in biological fluids at body temperature • Good film former (strong & flexible) • Concentrated (40%, w/w) aqueous solutions of gelatin are mobile liquids above 45 °C • This is extremely important during manufacture • Undergoes a solution-to-gel transition when cooled down
122
why does the API and the excipients need to be around the same size in capsules?
Particles with the same size have less tendency to separate in capsules where they could be spun or moved around.
123
Outline advantages of softgels.
1. Improved drug absorption: Reduction in variability for poorly soluble drugs 2. Patient compliance: Easy to swallow, taste-free 3. Safety of potent and cytotoxic drugs: Avoids dust, better operator safety 4. Oils and low melting point drugs: Overcomes problems that these drugs have as tablets 5. Dose uniformity for potent drugs: Liquid flow has higher precision than powder flow Liquids have better homogeneity than powder or granule mixtures 6. Product stability: protected from water and oxygen
124
outline dyes (colourant in both HGC and soft capsules)
• Dyes are defined as intensely coloured organic substances that impart colour to a substrate by selective absorption of light • Dyes are used in their water soluble form
125
outline esters of p-hydroxybenzoic acid (preservative for HGCs and soft capsules)
Mode of action: Interferes with cell membrane function, nucleic acid synthesis, and enzyme activity
126
outline opacifiers (colourant in both HGC and soft capsules)
• Work by scattering light due to a high refractive index • Titanium dioxide is used extensively as an opacifying agent • Protects from light and conceals the capsule content
127
outline osmotic release oral system.
• Includes a capsule that is coated with a semipermeable membrane • Allows water in but doesn’t allow drug out • The capsule contains a layer of osmotically active agent (push layer) and the drug formulation(s) layer(s) • Following ingestion, water is drawn into the push layer • This expands gradually, pushing drug out of the capsule • This system releases drug over a prolonged time, maintaining a more stable drug concentration in the blood
128
Outline pigments (colourant in both HGC and soft capsules)
• Pigments are defined as coloured particulate organic or inorganic solids which are usually insoluble in the substrate • They alter appearance by selective absorption and/or scattering of ligh
129
outline pulsincap capsules (pulsinated release)
• Includes an insoluble capsule body and a swellable/degradable plug, typically made of a hydrophilic polymer or lipid • The lag time is controlled by the plug, which is pushed away by swelling or erosion, allowing a pulse of drug release from the insoluble capsule
130
Outline sulphur dioxides (preservative for HGCs and soft capsules)
Mode of action: Antioxidant to prevent oxidative spoilage and is toxic to bacteria and fungi
131
outline the following methods of filling hard gelatin capsules (HGCs) with liquids / semi solids.
1. Piston filling • Volumetric dosing device • Prevention of leakage • Semi-solids are filled in the liquid state, where possible, either by heating for thermosoftening mixtures) or by stirring (for thixotropic mixtures) • After filling, these revert to the solid state 2. The effect of viscosity • The viscosity of the fluid affects the variability of the fill volume • Low viscosity: loss of liquid due to splashing from capsule during filling • High viscosity: difficult to pump and problems in transfer from nozzle to capsule body
132
outline why we use liquids / semi solids to fill HGCs
• Powder flow problems avoided • Avoidance of airborne particles • Improved fill weight accuracy • Convenient for formulations with a low melting point (e.g. oils) • Can improve solubility and absorption • Low dose/high potency drugs
133
what are the 3 categories of semi solids?
1. thermosoftening mixtures - filled in the molten state, when they can be pumped and filled , and subsequently solidify 2. Thixotropic mixtures - Thin upon shearing by mixing (low viscosity) - Form hard masses upon standing when shearing ceases (high viscosity) - Filled as fluids and semisolid during storage 3. Pastes
134
what is the difference between water miscible liquids and water immiscible liquids (as excipient selection in HGCs)
1. Water-miscible liquids Some PEGs/poloxamers • Vehicle dissolves/disperses readily in the GI fluids, liberating the drug either as a solution or fine dispersion • Depending upon aqueous solubility, this can aid rapid absorption 2. Water-immiscible liquids Lipophilic liquids, castor, olive, & sunflower oil • Release is followed by dispersion in the GI fluids • Drug may be present as an emulsion, fine suspension, or nano-emulsion • Well formulated systems to improve drug solubility will ensure that the drug does not precipitate in GI fluids
135
what is the issue with moisture content, what happens if moisture content is too high or too low? (HGCs)
The equilibrium moisture content refers to the amount of moisture that the capsule material will hold when it is in balance with the surrounding environmen. Moisture content too high: Capsule shell softens Moisture content too low: Capsule shell becomes brittle and can fracture Hygroscopicity is the ability of a substance to absorb moisture from its surroundings High levels of hygroscopic excipients must be avoided
136
what parameters need controlled in rotary die encapsulation method (manufacture of softgels)
• Temperature: Controls heat available for capsule seal formation • Timing: The timing of the dosing unit quantifies liquid fill matrix into the softgel • Pressure: The pressure exerted between the two rotary dies controls the softgel shape and final cut from the gel ribbon
137
outline mechanisms by how disintegrates work.
1. swelling - The disintegrants are hydrophilic non-soluble polymers with affinity to water. During uptake of water the polymer will swell to a degree depending on particle size, degree of cross-linking etc. 2. Wicking - effect of drawing water into a tablet 3. Recovery of energy of elastic deformation from compaction of the tablet - During tableting, particles may undergo elastic deformation but become interlocked before they can relax. As water penetrates the tablet matrix, these particles can relax and regain at least partially their original shape, thus forcing other particles away 4. Repulsion (mainly for starch) - particels in water diapers in such a way that they repel each other 5. Heat of wetting - The energy released during interaction with water heats up air entrapped in the tablet, which then expands 6. Interruption of bonding forces - During tabletting the particles interlock forming new bonds. These are interrupted when water is present thus releasing the particles
138
what are some properties of excipients?
- inert (except foe their intended characteristics) - be physiologically harmless - have comparable physical and chemical characteristics between batches - comply with certain microbiological standards depending on the administration route.
139
what are the two classes of disintegrants and what are some examples of each?
1. The traditional disintegrants • Native starches • Alginic acid • Ion exchange resins 2. The superdisintegrants • Sodium starch glycolate • Croscarmellose sodium • Crospovidone
140
What is a disintegrant?
• Disintegrant: Any solid, pharmaceutically acceptable material included in the formulation that acts to cause the tablet matrix to break up when the tablet comes into contact with aqueous media.
141
what is the function of diluents / fillers?
- they are employed to increase the mass of tablet, making manufacture more reliable and reproducible. - they must have good compression properties and be inexpensive
142
what is the main mechanism of action of super disinttegrants?
Swelling
143
what is the role of lubricants in tablets?
- Overmixing may impact disintegration and dissolution. - Mixing of disintegrant with lubricant should be avoided as it leads to formation of film on the surface of disintegrate reducing wettability. - Efficacy is increased if particle size is reduced
144
what do enteric coating tablets do?
protect the drug from the acidic environment in the stomach - enteric coating = gastro resistant
145
what is the difference between conventional compressed tablets and multiple compressed tablets?
conventional compressed tablets - designed to provide rapid disintigration multiple compressed tablets - Prepared by subjecting the fill material to more than a single compression - Each layer may contain a different medicinal agent, separate for reasons of chemical or physical incompatibility, staged drug release or simply for the unique appearance of the layered table
146
why do efferevescent tablets not need a disintegrates?
because it is a effervescence reaction - instead of a disintegrate, we need to use a carbonate salt eg citric acid and sodium bicarbonate.
147
see photo on phone from 24th january for exam question.
See photo from 24th January for answers
148
what are the 3 critical components in tablet coating (Coating composition)
3. Coating composition - plasticisers (added to coating to modify the physical properties of the polymer) - colourants (water soluble pigments) - solvent (to dissolve, disperse and convey the polymers and other additives)
149
what are the 3 critical components in tablet coating) (Coating process)
2. Coating process - coating equipment - parameters of coating process - spray application systems - automation coating process
150
what are the critical components in coating? (Tablet properties)
1. Tablet properties - tablet hardness (not less than 80N) - tablet friability (aim for 0.1%) - tablet core shape (as close to sphere as possible) - tablet porosity (increases force of attraction between coating) - Ingredients (do they interact with the coating)
151
what properties of tablet can be altered by coating of tablets?
- protection from light / moisture - protection from GI - taste masking - appearance masking - release profile
152
how do you calculate Q value?
Q value (%) = (mass of active substance released at given timepoint / total mass of active substance) x 100
153
what is disintegration? Why is it important?
- it is the assessment of the time taken for solid dosage forms to break down into smaller particles or granules It is important as: • Disintegration is an important pre-requisite to dissolution • It is vital for release of active ingredients • Quality control of disintegration is important to ensure tablets will break down in a predictable and timely manner • Ensures dosage forms will break down at the appropriate site within the body, e.g., gastro-resistant tablets
154
what is dissolution?
• A measurement of the rate and extent at which the active substance is release from a solid dosage form into solution
155
what is friability? Why is it important?
Friability is a measure of the physical resistance of the tablet to abrasion and breakage during production, packaging, transport, and handling before usage it is important: ensure tablets are durable enough to withstand the rigours of manufacturing, packaging, and handling without crumbling or breaking apar
156
what is hardness? Why is it important?
• The force required to break the tablet (tablet breaking force) - why is it important? - An extremely hard tablet could indicate excessive bonding potential between active ingredients and excipients, which can prevent proper dissolution of the tablet needed for an accurate dosage - A softer tablet could be a result of weak bonding and may lead to premature disintegration when ingested by the patient - Patient acceptability and compliance
157
what is uniformity of content?
• The test for uniformity of content is based on the assay of the individual contents of active substance(s) • It is determined for a number of single-dose units to determine whether the individual contents are within limits set with reference to the average content of the sample - it is important as it is critical to know how much of the active substance is in each dosage unit
158
what is uniformity of weight?
- it is ensuring each dosage form is consistent in weight - it is important that we do this as uniformity off mass is linked to the dosage the patients receive
159
outine the considerations to take for the following when considering development of an ocular solution based product. ➡ Solubility, ➡ Osmolarity, ➡ Vehicle Viscosity, ➡ Stabilisers, ➡ Antimicrobial Preservatives
➡ Solubility = choice of salt in the formulation, ➡ Osmolarity = products may be tolerated over a fairly wide range of tonicity. ➡ Vehicle Viscosity = increase in vehicle viscosity = the residence time in the eye ➡ Stabilisers = drugs that are prone to degradation need these ➡ Antimicrobial Preservatives = for sterility
160
outline ointments for delivery of drugs to the eye
๏ Eye ointments are sterile semi-solid preparations intended for application to the conjunctiva. ๏ Increased contact time & better bioavailability compared to solutions. ๏ Night-time application, ๏ Poor patients compliance - often cause blurred vision.
161
Typically how much of a drug which we apply topically to the eye will actually permeate into the cornea?
<10% (typically 1%)
162
what are some advantages and disadvantages of solutions in treatment of eye conditions.
advantage - high solution concentration can be achieved - good dose uniformity and better bioavailability - easy to manufacture disadvantage - rapidly drained from the eye (makes you blink more)
163
What are sone advantages of water-based gels in treating eye conditions?
Advantage - ease of administration, - improved patient compliance
164
what are the 4 types of contact lens?
1. hard lens 2. Semi hard 3. Soft lens 4. Soft continuous wear lens
165
what are the physiological barriers of the eye from drugs?
- conjunctiva = microvilli layer - cornea = tight junctions - sclera - tear production/blinking - protein binding
166
what influence does solubility of a topical eye drug have on its uptake? (Diffusion vs carrier mediated) also logP for corneal permeation?
For most topically applied drugs, 1. Passive diffusion along the concentration gradient, either transcellularly or paracellularly, is the main permeation mechanism across the cornea. 2. Carrier-mediated active transport mechanism is also indicated. LIPOPHILIC DRUGS TEND TO FAVOUR THE TRANSCELLULAR ROUTE HYDROPHILIC DRUGS TEND TO FAVOUR THE PARACELLULAR ROUTE - therefore drugs that have both lipophilic and hydrophilic properties tend to permeate most effectively - logP = 2/3 fot corneal permeation
167
what is the 4 routes which we can delivery drugs to the eye via?
- Topical administration - Systemic (parenteral) administration - Oral administration - Periocular & intravitreal administration
168
what is the optimal charge for permeation, wha is the optimal molecular size?
- unionised form usually permeates the lipid membranes more easily than the ionised form - corneal epithelium is negatively charged - charged cationic drugs permeate more easily through the cornea than anionic drugs do - molecular size <500 daltons
169
when can you consider suspension for delivering drugs to the eye? How do they work?
Aqueous or oily suspension eye-drop formulations may be considered for drugs that are poorly water soluble, or because of poor aqueous drug stability Prolong the residence time of drug particles in the eye, allowing time for dissolution in the tears and an increase in ocular bioavailability
170
Explain why the equation on slide 6 is pseudo 0 order reaction?
as the g/ml (2.5/100) is much higher than the solubility of of ampicillin means we have a suspension. Therefore, the amount of ampicillin in solution is always constant. And because the OH- is buffered (remained constant) therefore it is a pseudo 0 order reaction
171
explain why the reaction on slide 5 is a pseudo 1st order reaction with respect to ampicilin?
as the solution is buffered, this means that the concentration of OH- is kept constant, hence the only thing that is “changing” is the ampicillin. Hence it is pseudo 1st order
172
look at karl lectures from level 1 to see the 1st order rate equation and 0 order rate equation and also the units for the rate constant
173
what does pseudo mean?
Apparent Therefore pseudo order reaction = apparent order reaction
174
why can the sucrose reaction on slde 3 be considered to be pseudo first order?
Chemically speaking, the equation is first order with respect to sucrose, H20 and H30+. So we add these up to get a reaction rate of 3. This can be considered psuedo first order because the sucrose is dissolved in H20, hence the H20 can always be in excess. Also because we have equal amounts of H30+ on each side then we can essentially forget about it. Hence, the reaction is pseudo with respect to sucrose
175
you do not need to know / will be asked to reproduce the integration etc
176
for specific base catalysis and specific acid catalysis, what is the slope of a plot of log k vs pH?
- specific acid catalysis = straight line with slope of -1 - specific base catalysis = straight line with slope of +1
177
hoe do you overcome acid specific catalysis?
- formulate it at close to neutral pH
178
how would you design an experiment to see whether a formulated pharmaceutical product undergoes acid or base specific catalysis?
design drug assay, changing ph values over range, measure drug concentraion at time points, get log K and plot log K vs Ph. If its -1 it is acid specific and +1 is base specific.
179
no flashcards for this see lecture slides
180
outline specific base catalysis
181
the maths stuff for this lecture could be asked on the exam
182
what does a plot of logk vs pH look like for specific acid-catalysis look like and how do you get to this?
183
what is specific acid-base catalysis? When does a reaction subject to it?
when the rate law for the degradation of a drug contains a term involving either [H+] or [OH-], the reaction is subject to acid-base catalysis.
184
what is the reaction equation for specific acid-base catalysis and define the terms
k = kH[H+] + kOH[OH-] + k0 Where k is the apparent 1st order rate constant, kH is the specific acid rate constant and KOH is the specific base rate constant, k0 represents the rate constant for the pH independent reaction, ie catalysis by the solvent (usually water)
185
Outline the units of radioactivity.
Basic units are the curie and the becquerel
186
outline what is meant by the roentgen, the rad and the rem.
roentgen: a unit for measuring the amount of gamma radiation rad: a unit for measuring the absorbed energy from radiation rem: a unit for measuring biological damage from radiation
187
what are gamma emitters?
this is often a secondary process that occurs after initial decay of B emissions, each isotope emits y rays of a distinct energy. This can be measured for identification of the isotope. - they will get back to the ground state by emitting energy, which will release a package of energy as discrete photon energy
188
what are the two forms that a beta particle can exist as? Outline some points around this.
1. electron formed by B- decay 2. positron formed by B+ decay - these are the most commonly used radioisotopes in biochemistry - emitted in a range of energies form a given isotope (energy is characteristic of an isotope) - The anti neutrino is an almost massless particle that carries away some of the energy from the decay process - These particles can travel 15 to 1600 cm in air - They can be stopped by low atomic number materials, such as plastics - Particles emitted from tritium sources have insufficient energy to penetrate outer dead layer of skin
189
what is alpha emission? Outline features of it.
1. Emitted particle is a nucleus of helium eg 2 neutrons and 2 protons It is: • heavy • slow moving • densely ionising • causes considerable biological damage • unsuitable for tracer work • travels a few cm in air and can be stopped by paper
190
what is the difference between radioactivity and nuclear stability?
1. Radioactivity - - results from spontaneous nuclear disintegration arises from unstable isotopes. 2. Nuclear stability - depends on the ratio of neutrons to protons - gives rise to spontaneous decay with emission of particles.
191
what is the equation for % efficiency?
= ((observed cpm - background cpm) / actual dpm) x 100
192
what is the gray? unit of radiation.
The gray describes amount of energy transferred by radiation to an object An absorbed dose of 1 gray is equal to the absorption of 1 joule of radiation energy by 1 kilogram of matter.
193
what is the sievert? Unit of radiation
The sievert is the correct unit to use when you wish to monitor the biological danger of radiation the difference between the rem and the sievert is a proportionality factor: 100 rem equal 1 sievert.
194
what rate is disintegration and what is the rate of decay proportional to?
Spontaneous disintegration is a first-order process.
195
Rate of radioactive decay is proportional to the number of atoms present
196
197
Give an example of a bone seeking agent and 3 advantages of it.
99mTc-methylene diphosphonate (MDP) Bone Scan • Detects lesions long before skeletal x-ray • Fast blood clearance • High skeletal affinity
198
outline gamma scintigraphy (radio pharmacy imaging)?
the RP emits gamma radiation: - Detected by a gamma camera or SPECT (single-photon emission computed tomography) - Important that the energy of the decaying gamma ray photon is within the specific energy window of the camera (generally 100-250 keV) - Gamma decay energies outside of this range may produce low-quality images
199
outline positron emission tomography (PET) (detection of radiopharmaeuticals)
• Positron decay results in the emission of two 511 keV gamma ray photons 180oC apart • Detected by a PET camera • PET scanners contain a circular array of detectors designed to specifically detect the 511 keV photons emitted in opposite directions
200
outline the principle of brain imaging. And also some indications.
• Water-soluble radiopharmaceuticals are excluded from normal brain due to intact BBB • To screen patients for the presence of primary tumours • To detect cerebral metastases • To evaluate patients with cerebrovascular disease • To detect intracranial injury • To study patients with intracranial disease
201
What are some used of thyroid imaging agents, and an example of an agent used.
• To image solitary or multiple thyroid nodules • To assess thyroid size and function • Aid in the management of thyroid cancer • Typically radioactive iodine- based (usually oral 123I NaI)
202
what are the mechanisms of localisation in radiopharmacy?
• Active transport • Simple diffusion • Capillary blockade • Phagocytosis
203
what factors are important to consider when choosing a radiopharmaceutical for diagnostics?
• Minimum half-life • Mode of decay • Cost and availability • Physical properties • Organ/tissue specificity
204
what is an example of a metabolism-based radiopharmaceutical? Outline points about this.
2-[18F]fluoro-2-deoxyglucose (FDG) • Rapidly distributes to all organs of the body • PET images yield apparent data on glucose utilisation and hence metabolic activity o Distinguish viable, but functionally impaired, ischemic myocardium from nonviable (infarcted) myocardium o Indicated for neurological and psychiatric disorders, especially associated with foci of epileptic seizures o Can also differentiate benign from malignant tissue
205
what is important about the half-life? What should it be?
Should be: • Long enough to carry out the desired chemistry to synthesise the radiopharmaceutical • Long enough to accumulate in the target tissue while clearing non-target organs - it must minimise radiation dose to the patient - it must range from 2 mins to several days
206
what is the therapeutic application of radiopharmaceutical?
Primarily directed against cancerous tissues utilising “the concept of tissue radiosensitivity” i.e. rapidly dividing cells are more radiosensitive
207
what radio isotopes are used for the assessment of - myocardial ischemia - myocardial necrosis - myocardial inflammation
- myocardial ischemia = TI chroride / TI isonitriles - myocardial necrosis = In antimyosin / Tc glucarate - myocardial inflammation = ln leukocytes
208
how does a vaccine work?
1. antigens added to bloodstream (vaccine) 2. Antigen presenting cells capture / ingest antigen, displaying pieces of it on its surface 3. These activate T helper cells, which trigger killer T cells and B cells 4. These cells then target the antigen 5. Therefore if the antigen is presented again then there is memory killer T cells and memory b cells
209
make sure to know an example of each type of vaccine to be able to say if asked a question.
210
what are 3 types of subunit vacicines. What is each made using?
1. Conjugate vaccines • Made using the polysaccharides, or sugars, that form the outer coating of many bacteria 2. Toxoid vaccines • Made using inactivated version of bacteria releasing toxins 3. Recombinant protein vaccines • made using bacterial or yeast cells to manufacture the vaccine
211
what are the 3 groups of types of vaccines?
- whole pathogen vaccines (• Live attenuated (weakned ) / • Inactivated) - sub unit vaccines - nucleic acid vaccines - viral vectores vaccines
212
what is a live attenuated vaccine?
• Obtained from live pathogenic organisms • Pathogenic organisms are treated to become attenuated (or weakened) – lose capacity to induce full-blown disease but retain immunogenicity • Pathogenic organism is attenuated by introducing it into a species in which it does not replicate well or forcing it to replicate repeatedly in tissue culture • Activates T-killer cells
213
what is a nucleic acid vaccine?
• Unlike other vaccines, these vaccines do not supply the protein antigen to the body • Provide the genetic material (DNA or RNA) of specific antigens to develop immunity • Nucleic Acid Vaccines include: DNA and RNA Vaccines eg covid vaccine was RNA
214
what is a sub unit vaccine?
• One or more antigenic fragments of the pathogen (e.g. proteins, polysaccharides, capsid) are used to stimulate the immune response • Antigens may be obtained through recombinant protein expression, production in yeast cells or bacteria or extraction from infected cells • Generally require addition of an adjuvant
215
what is a vaccine?
• A vaccine is a biological preparation • Typically contains weakened or inactive parts of a particular disease-causing pathogen
216
what is a viral vectored vaccine?
• Like nucleic acid vaccines, these vaccines do not supply the protein antigen to the body • Provide the genetic material (DNA or RNA) of the antigen to host cells using a safe virus • In turn, the cells produce the antigen which stimulates an immune response • Viral vectored vaccines include: Replicating and non-replicating vaccines
217
what is herd immunity?
• Refers to indirect protection of a community of people from a disease by immunising a critical proportion of that population
218
what is the difference between the innate and adaptive immune system?
innate: • First line of defense • Responds quickly to invaders adaptive: • It has been ‘educated’ • Second line of defense • Slower response
219
what is the R0 value?
• Term that describes how contagious/transmissible an infection disease is • R0 = Basic Reproduction Number • Average number of secondary cases arising from a primary infection case in an entirely susceptible population How many people does one infected person averagely infect?
220
aside from the active ingredient (antigen) what is the main ingredient in a vaccine?
Sterile water
221
outline the process of intramuscular vaccine administration.
• Identify the correct site for IM injection • Stretch the skin at the site • Insert the needle at a 90° angle • Depress the plunger • Gently remove the needle • Apply light pressure if bleeding occurs
222
outline what the main ingredients of a vaccine are.
- antigen - adjuvants - preservatives - stabiliser - surfactants - residuals - diluent
223
what are the three phases of clinical trials?
1. Phase 1 - Tested on a small number of healthy volunteers - assesses safety - determines the right dosage 2. Phase 2 - Tested on several hundred volunteers - Volunteers in this phase have the same characteristics (age, sex) as the people for whom the vaccine is intended. - assesses safety - determines correct dosage - confirms vaccine generates an immune response 3. Phase 3 - Tested on thousands of volunteers - determines efficiency of vaccine - assesses safety in large group
224
what is the cold chain?
describes the recommended cold temperature conditions for a vaccine according to the manufacture during transport and storage until the point of administration • Vaccines must be kept in the cold chain from the manufacturer to some of the most remote places on earth • Vaccines are highly susceptible to environmental factors
225
what is the role of the adjuvant in a vaccine?
- Improves the immune response to the vaccine - Examples of adjuvants: Aluminium salts (like aluminium phosphate, aluminium hydroxide or potassium aluminium sulphate)
226
what is the role of the antigen in a vaccine?
- Active component or contains ‘blueprint’ to make the active component - Generates immune response
227
what is the role of the diluent in a vaccine?
- Dilute a vaccine to the correct concentration immediately prior to use
228
what is the role of the preservative in a vaccine?
- Prevent the vaccine from becoming contaminated once the vial has been opened - Not all vaccines contain preservatives (not needed)
229
what is the role of the residuals in a vaccine?
- Tiny amounts of various substances used during manufacturing or production of vaccines
230
what is the role of the stabiliser in a vaccine?
- Prevent chemical reactions from occurring within the vaccine - Keep the vaccine components from sticking to the vaccine vial
231
what is the role of the surfactant in a vaccine?
- Keep all the ingredients in the vaccine blended together - Prevent settling and clumping of elements that are in the liquid form of the vaccine
232
where should vaccines be administered?
• Injected into the deltoid muscle in the upper arm (2 fingers down) • If insufficient muscle mass in the area of the deltoid or a particular reason the deltoid muscle is otherwise unsuitable, vaccine should be injected into the vastus lateralis muscle in the anterolateral aspect of the thigh
233
outline the 4 grades of manufacture of sterile medicinal products.
grade a = the local zone for high risk operations. These conditions are provided by laminar air flow: providing a homogeneous air speed of 0.45m/s +- 20% at the working station. grade b = this provides the background environment for grade A zone items needing aseptic preparation and filling grade c/d = areas graded C and D are used for performing less critical tasks that are carried out during less critical stages in the manufacturing process.
234
outline the overkill method (sterilisation process)
(most popular) - start with an unrealistic bioburden, find out a D value form this (usually spores of thermophile) - then complete for a total of 12 D values, which will kill all ‘normal bacteria”
235
outline the sterility assurance level? (Sterilization process)
- sterility is an absolute concept (either sterile or not) - it is The likelihood of a product being sterile (free from microorganisms) can be expressed in terms of the probability of a microorganism surviving the treatment process. - this gives rise to the concept of Sterility Assurance Level (SAL)
236
what 2 factors influence the time taken to sterilise a product?
- (N0) initial level of contamination (the bioburden) - k - the rate of kill, which is dependent on the temperature and the thermal resistance of the organism
237
What are the 3 stages of process validation?
- Stage 1 –PROCESS DESIGN: The commercial process is defined during this stage based on knowledge gained through development and scale-up activities. - Stage 2 – PRCESS 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.
238
what are the 3 stages of process validation?
• 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.
239
What are the 3 things important in good manufacturing practice?
- personell - process - environment
240
what do you need to consider when choosing a sterilisation process?
- knowledge of the lethality of the chosen sterilization process (D value)
241
what does validation of a sterilisation process require knowledge of?
• Lethality of the process • Effects of changing sterilization conditions
242
what is a phase transition?
when a gas at its phase boundary temperature interacts with a colder object, it undergoes phase transition (think of a kettle gaseous water hits cold air around it and recondneses = steam)
243
what is latent heat (energy)?
this is hidden heat - we don’t see a temperature increase despite adding heat (eg water @100C - can keep using a bunsen burner but temp won’t rise)
244
what is parametric release?
Parametric release is a system of release that gives the assurance that the product is of the intended quality based on information collected during the manufacturing process. - based on evidence of successful validation of the manufacturing process
245
what is process validation?
process validation is defined as the collection and evaluation of data, from the process design stage throughout production, which establishes scientific evidence that a process is capable of consistently delivering quality products. Process validation is a requirement of current Good Manufacturing Practices
246
what is sensible heat (energy)
is the amount of heat that, when added or removed from a substance, causes a change in its temperature but does not cause a phase change (eg raising water from 40C to 60C)
247
what is the D value?
time (mins) taken to reduce the population size by 90%
248
what is the Sterility Assurance Level (SAL)?
the degree of assurance with which the process in question renders the population of items sterile
249
what is the Z value?
increase in temperature required to reduce the D value for an organism by 90% reduction
250
what is thermal death time?
time required to kill a population of the target microorganism at a given temperature
251
what products can parametric release only be applied to?
Parametric release can only be applied to products terminally sterilised in their final containers
252
What’s the definition of F value and what is it used for?
- F is defined as the equivalent time at temperature (T) is delivered to a container or unit of product for the purpose of sterilization. - Permits comparison of lethal effects of heating at any temperature to prescribed conditions (121oC)
253
Use lecture slides and do practice questions
254
list methods used to reduce particle size.
- cutting methods - compression methods - impact methods - combination impact and attrition
255
outline combination impact and attrition in size reduction (High pressure homogeniser)
• Sample forced to pass through a very small gap at high pressures (500-2500 bar) • Impact and attrition lead to micro/nanonisation • The sample can recirculate to the homogenisation chamber
256
Outline combination impact and attrition method in size reduction (Media mill)
• Small beads (0.1 – 5 mm) are the milling media • Impact and attrition lead to micro/nanonisation • The sample recirculates to the milling chamber
257
outline cutting method of size reduction.
• Consists of a series of knives attached to a horizontal rotor • Knives act against a series of stationary knives attached to the mill casing. • Size reduction occurs by fracture of particles between the two sets of knives, which have a clearance of a few millimetres. • A screen is fitted in the base of the mill casing; thus it is self-classifying.
258
outline cutting methods in size reduction/
• Consists of a series of knives attached to a horizontal rotor • Size reduction occurs by fracture of particles between the two sets of knives, which have a clearance of a few millimetres.
259
outline direct compression.
steps involved are: • Mixing of excipients and drug (planetary bowl/rotating drum/high- speed) • Compression of powders into tablet
260
outline dry granulation.
• Involves production and subsequent compression of granules into final dosage form • No solvent is required • Two principle methods 1. Slugging - double compression technique, mix drugs, form large tablet, mill, compress 2. Roller Compaction - mix drugs, compress powder between two rollers, mill the ribbon produced, compress into tablets
261
outline wet granulation.
particles interact to form aggregates Two primary stages involved: - Particle-particle interactions facilitated by formation of liquid bridges - Particle-particle interactions facilitated by formation of solid bridges.
262
outline why particle-particle interactions occur during drug granulation.
1. Electrostatic forces - Play a role in initial cohesive interactions but not so in granule strength. 2. Van der Waals interactions - major role and increase in magnitude as particle distance decreases 3. Melting of components - Granules exposed to high shear rates that may result in partial melting. Upon cooling (solidification) increased interactions between adjacent particles occur
263
what 3 factors contribute to an improved bioavailability?
- Increased dissolution rate - Increased saturation concentration - Enhanced bioadhesion
264
what are the different ways that wet granulation can be carried out?
• Blending & Oscillating Granulator • High Speed Mixer/Granulator • Fluidised Bed Granulatior • Extrusion Granulator
265
what benefits do milling bring about?
• Improved Drug Dissolution • Uniformity of Dosage Forms • Enhanced Mixing and Homogenization • Tailoring Drug Release Profiles • Micro and Nanosizing • Reduced Agglomeration
266
what do powders need to have to be mixed? How do we get this?
powders need to have the same particle size, this is done via milling
267
what does milling aim to do?
• It allows the manipulation of physical properties of active pharmaceutical ingredients (APIs), and excipients. • By reducing particle size, milling enhances the dissolution rate, bioavailability, and overall performance of drugs in various dosage forms. • The primary objective is to reduce the size
268
what does the binder do?
helps the powder stick together during compression
269
what does the lubricant do?
ensures that when the powder is compressed into a tablet it doesn’t stick within the metal die
270
what does unit operations mean?
individual steps or processes involved in the manufacturing of pharmaceutical products.
271
What is a negative mixtures? (Complete segregation)
Negative mixing occurs in a two-phase system where the phases differ in density and will separate unless continuously agitated. Example: Oil and water.
272
what is a neutral mixture? (Random mix)
Neutral mixing occurs when neither mixing nor separation of the system occurs unless it is acted upon by a system of forces
273
what is a positive mixture? (Ideal/perfect mix)
Positive mixing applies to systems that given time, would spontaneously and completely mix. Example: Two gases or miscible liquids.
274
what is Fluidised Hot-Melt Granulation (FHMG)?
bringing particles together - Melt agglomeration obtained through addition of a solid binder that melts during the granulation processing (50-80°C) - Mechanism similar to granulation with liquid addition - No need for drying
275
how do you classify between small volume parenteral and large volume parenteral?
- Small volume Parenterals (SVP) (100ml or less) - Large volume Parenterals (LVP) (up to 500ml)
276
list some advantages of parenteral administration?
- Local effects can be achieved where desirable – anesthesiology - Formulations can permit prolonged drug release profiles  Depot injections  Long-acting steroids  Long-acting penicillins by deep IM injection - Provides a means of correcting serious disturbances of fluid and electrolytes - When food cannot be swallowed or taken orally, total parenteral nutrition can be tailored to meet the individuals exact nutritional needs
277
outline water for injection.
- Clear, colourless and odourless (pH 5 - 7) - Purity specifications limit - Cl-, Ca2+, SO42-, NH3, CO2, heavy metals and oxidisible substances total amount of dissolved solids < 10 ppm - Must be used within 24h unless sterilised, sealed in ampoules or containers
278
what % gram moleular concentration is an isotonic solution?
0.0003
279
what are disadvantages of parenteral administration?
- Requires aseptic technique - Skill of administration required - Pain on administration - Rapid development of allergic reactions in sensitive individuals - Difficult to reverse effects of drugs given parenterally, even immediately after administration - Inherent requirements (sterility, freedom from particulate matter & pyrogens, stability) requires understanding from all personnel who manufacture or administer the formulation
280
what are requirements of large volume parenterals?
- Sterile - Non-pyrogenic - Free of Particulate Matter - Packaged as single dose containers
281
what injections should preservatives not be added to?
- Intrathecal, intracisternal and peridural (epidural) use (CSF injection) - Intracardiac or intraocular injections - Injections in which a single dose is greater than 15mL, unless the BP monograph specifically allows the presence of a preservative
282
what is a colligative property?
When a non-volatile solute is dissolved in a solvent certain properties of the resultant solution are largely independent of the nature of the solute and are determined by the concentration of the solute. Most important one for pharmaceutical pot is osmotic pressure
283
what is a hypertonic solution?
- in a hypertonic solution (osmotic pressure greater than blood serum), water will pass out of the cell - the cell shrinks and the cell wall appears crenulated
284
What is a hypotonic solution?
- In a hypotonic solution (osmotic pressure less than blood serum), water will pass into the cell until equilibrium is established - the cell swells rapidly and bursts
285
What is a preservative?
Antimicrobial preservative agents serve to maintain sterility of the product during shelf life and use
286
what is a pyrogen?
a substance that induces fever - they are derived mainly from gram negative bacteria
287
what is an isotonic solution?
- Solution where the osmotic pressure is the same as the blood serum - This is what all parenteral systems need to be
288
what is the intradermal route?
- drug injected into superficial layer of skin - absorption is slow - generally reserved for diagnostic tests - small volumes around 0.1 - 0.2mL
289
what is the intramuscular route?
- Volume circa 2 mL (up to 5 mL gluteal administration) - Typical sites include the shoulder (deltoid), thigh and buttock (gluteal medials) - Very suitable for aqueous and oily solutions and suspensions which are irritant by the SC route - More rapid absorption than SC - can be modified by formulation type (oily liquid, suspension etc.)
290
What is the intravenous route?
- Large or small volumes (up to 500mL) can be administered to the veins for rapid effect - Formulations relatively simple - active drug, other ingredients (TPN components, preservatives etc.) and a solvent system (preferably aqueous) - Results are rapid and predictable but potentially dangerous (no retreat following administration) - Irritant drugs rapidly diluted in blood - Number & location of vein = accessibility
291
what is the osmotic pressure?
Simply the minimum pressure needed to prevent osmosis. - The force per unit area, or pressure, required to prevent the passage of water through a selectively permeable membrane and into a solution of greater concentration is equivalent to the osmotic pressure of the solution
292
what is the subcutaneous route?
- Drug injected into loose subcutaneous tissue beneath skin - Volume circa 1 mL - Usually administered in the upper arm or thigh - Popular route of administration (e.g. insulin) - More rapid onset of action than intradermal
293
whats the acceptable range of pH?
36991
294
whats the difference between enteral and parenteral?
enteral = administered via the gut Parenteral = administered by bypassing the gut
295
whats the difference between suspensions and emulsions?
suspension = dispersion of drug in one continuous phase Emulsion = disperse systems in which one immiscible liquid is dispersed in the other
296
how do substituents have an effect on solubility?
the effects are mainly due to - effect on the properties of the solid or liquid, e.g. molecular cohesion - effect of the substituent on its interaction with water molecules ie whether its hydrophobic or hydrophilic - hydrophobic will have better solubility
297
how does drugs acidity, basicity, or amphoteric effect there solubility/
- acidic drugs are less soluble in acidic solution (over abundance of protons) - basic drugs are more soluble in acidic solutions (as it gains a proton) - For amphoteric drugs, it is observed that the zwitterion has the lowest solubility
298
how does the location of substituents on a benzene ring effect solubility?
• The relative low solubility of the para compound is due to the greater stability of its crystalline state • In the ortho state, intramolecular hydrogen bonding in aqueous solutions may occur, therefore decreasing the interaction of the OH group with water
299
Outline cyclodextrins
• Modified starches • Glucopyranose units form a ring : 6 unit ring : 7 unit ring : 8 unit ring • The ring is cylindrical with a hydrophilic outer surface and a hydrophobic interior • Appropriately sized lipophilic molecules may be housed in the central portion
300
what are 6 excipients commonly saw in liquids, what does each do and an example/
1. Vehicle = act as a solvent or carrier 2. Preservative = control microbial growth eg parabens 3. Sweetening agents = add flavour eg sucrose 4. Viscosity = optimise pour ability eg modification of conc of sugar or hydrophilic polymers 5. Flavours = salt, bitter, sweet, sour 6. Colours = provide colour
301
what are some advantages of pharmaceutical liquids?
• Ease of administration compared with solid dosage forms • Increased rate of drug absorption from solutions • Taste of medicines may be effectively masked
302
what are some disadvantages of pharmaceutical liquids?
- Difficult to formulate drugs that possess poor chemical stability - Difficult to formulate liquids for drugs that possess poor solubility (Specialist techniques may be required to enhance solubility) - Require specialist formulation strategies (Taste masking, Appearance, Viscosity)
303
what effect does the melting point of a drug have on its solubility?
drugs with a higher melting point will have poorer solubility than that of a drug with a lower melting point
304
what is a cosolvent
a water miscible liquid that has less hydrophilic properties than water
305
what is hydrotropy?
• Salts with large anions or cations that are soluble in water may increase the solubility of non-electrolytes - Due to the dissolution of the solute in oriented clusters of the hydrotrope - Large concentrations of hydrotrope required (20-50%)
306
what is the dielectric constant?
– Maximum solubility is achieved in a particular solvent systems at a defined dielectric constant – The dielectric constant is equal to the capacitance of the condenser filled with some reference material (Cx) divided by the standard (Co)
307
what is the solubility parameter?
– Thermodynamic measure of the cohesive forces that exist in a substance – Maximum solubility is achieved whenever the solubility parameters of the solvent and solute are identical
308
whats the advantage of nonionic surfactants?
they are far less toxic than ionic surfactants
309
European Medicines Agency states that choice of preservative is supported by the following:
– Why the preservative is included – Proof of its effectiveness – Methods and standards for its control in the product – Labelling details on the product – Relevant safety information
310
for preservatives to be effective, what properties must they have?
- preservation must be available in the aqueous phase - they need to be unionised (if they are ionised them they will cross the membrane of microorganism and therefore undergoing a dilution event = overall available amount of preservative available decreased.
311
outlibe phenols as preservatives
Widely used as disinfectants and preservatives • 0.5% solution of phenol will kill most vegetative bacteria • Activity markedly diminished by dilution and presence of organic matter • Numerous incompatibilities • Odour, toxicity and volatility issues
312
outline circumstances which preservative efficacy and avilability may be altered.
- Partitioning into oil/aqueous or biophase - Ionic status & pH - Adsorption & Permeation to container & Closure - Temperature & Surfactant - Status in tablets, suspensions, gels, creams, nasal and eye drops - Physical or Chemical interaction with other formulation components - Microbial Challenge (dilution of the preservative as microorganisms take it in the overall concentration available reduces)
313
outline cresol as preservative
• Mixture of o-, m- and p-methylphenol • Poor solubility • 0.3 – 0.5% effective concentration ranges in suitable formulations • Similar activity range to phenol but less caustic and less poisonous • Activity not reduced in presence of organic matter
314
outline parabens as preservative.
- Series of alkyl esters of p-hydroxybenzoicacid • Less readily ionised than parent acid • pKa values around 8.0 -8.5 • Exhibit good preservative activity even at pH 7-8 • Mixtures of esters to protect the various phases in a multiphase formulation
315
outline preservative efficacy testing
- The test may be conducted in either five original market containers or in five sterile, capped bacteriological containers into which a sufficient volume of the formulation has been transferred - Each container is inoculated with one of the prepared and standardised inoculum to give a final concentration of microorganisms in the test formulations of 10^5 – 10^6 cfu per mL or per g of the product, - mixed thoroughly to ensure homogeneous distribution of microorganims throughout the formulation - Incubate the inoculated containers at 22.5 +/-2.5C (USP)/20-25C protected from light (BP) and sample each container at appropriate intervals, over a maximum of 28 days, according to the type of product. - BP suggests 1 mL or 1 g sample sizes taken at the appropriate intervals, starting from zero hour - The number of viable organisms in each sample is determined by plate count (USP) or by plate count or membrane filtration (BP) - Any changes to the appearance of the product should also be recorded - its important to quench the reacrtion at the time points so that we dont get a better result than we actually did
316
outline the mechanism of action of weak acid preservatives.
• Lipophilic Unionised species diffuses across membrane • Encounter cytoplasmic pH near neutrality • Forced to Dissociate to charged ions • Charged ions cannot return across membrane – Anions concentrate in cytoplasm • Dissociation of acid derives proton • Cytoplasm acidification • Growth is Inhibited
317
what 4 ways does the acidification of cytoplasm inhibit growth
• Inhibition of glycolysis • Inhibition of folic acid synthesis • Prevents Active Transport • Interference/ Inhibition of signal transduction
318
what characteristics must every pharmaceutical product have?
• Elegant (must look like how its meant to) • Efficacious • Stable • Safe All throughout their intended life
319
what does Aw stand for? What is the use in this?
product water activity - In complex formulations, microorganisms must compete for water against other components of the formulation which interact with water - Certain products may be protected by manipulation of Aw (ready availability of water to contaminants), to values below those required for growth it can be lowered to give preservation effects by addition of water binding low molecular weight solutes
320
what effects does suspended solids have in a solution?
they completely wipe out any preservative
321
what is an objectional organism
An objectionable organism is a microorganism that, if present in a product or manufacturing environment, poses a risk to product quality, safety, or compliance with regulatory standards.
322
what is the definition of a preservative?
Substances added to pharmaceutical formulations/medicines to prevent microbial spoilage, retard deterioration, and restrain organismic growth to low levels
323
what is the validation process of preservation efficacy testing?
it must demonstrate: – Adequate mixing of the inoculum with the product – Effectiveness of the inactivator or method used to neutralize preservative activity within the sample – Lack of microbiological toxicity of the inactivator which would interfere with the test data and; – Ability of the procedure to bring about the necessary reductions in viable count of the starting inoculum within the formulation/product.
324
when must preservatives be added to formulation?
by law they need to be added to any formulation which is to be used more than once from the same container
325
why is pH control ineffective as a preservation technique?
pH dictates the type of spoilage that occurs Eg • Low pH fruit juice-flavoured medicines inhibit bacterial growth but favour fungal growth • Higher pH anatacids etc. favour growth of pseudomonads