What is cleaning validation?

Cleaning validation is a process to ensure that equipment cleaning procedures are removing residues to predetermined levels of acceptability. Although “equipment cleaning” is part of current Good Manufacturing Practice requirements the term “cleaning validation” was not popular until late 1980s. The need for a systematic approach to proving the effectiveness of all the cleaning procedures was achieved in 1993 with a revised Food and Drug Administration Inspection Guide on Cleaning Validation.

Pharmaceutical products and active pharmaceutical ingredients (APIs) can be contaminated by other pharmaceutical products, by cleaning agents, micro-organisms or by other material (e.g. air-borne particles, dust, lubricants). Further sources of contamination might be raw materials, intermediates, auxiliaries, etc.

In many cases, the same equipment may be used for processing different products. To avoid contamination of the following product, adequate cleaning procedures are essential.

High risk products, such as penicillins, are a major concern in the cleaning validation field. Sensitive sampling methods require development and must be applicable to each specific piece of equipment used. Due to the possibility of inter-product contamination, highly sensitive analytical methods such as Liquid Chromatography are required for trace level analysis.

These detection methods should be specific for the target analyte, sensitive for trace and ultra-trace analysis, and be sufficiently able to separate and quantify the target analyte from potential interference. Currently there are two sampling methods in use.

The direct method, incorporating a swabbing material, is favoured over the indirect rinsate analysis approach. However caution must be taken when choosing the correct swab, as factors such as recovery, background contribution and particle generation can hinder residue determinations. Therefore each should be evaluated independently and an overall correction factor applied to the swab. Swab recoveries may be determined using spiking studies incorporating coupons of equipment surfaces.
Rinsate analysis is a useful sampling tool for equipment such as blenders and reaction vessels. The theory is that by analysing an aliquot of rinse water, the total quantity of analyte residue can be estimated. This method however assumes that the residue is uniformly removed from the equipment and also presumes that if the rinsate is clean then the equipment is clean.

Probably the most important aspect of cleaning validation programmes is establishing predetermined levels of acceptability. In order to establish Acceptable Residue Limits (ARL), various product and equipment attributes are evaluated. This leads to wide variations in ARL values between different product trains and different manufacturing facilities. The most important aspect is therefore proving that the ARL values determined in the cleaning validation programme have been established using a sound scientific rationale.

1 Like

Very informative

Thanks

[dear sir,
I am Arutla,
please give me how to use this thread?

Hi Arutla,

What do you mean, do you want to add to this thread otr create a new one ?

Dear Sir,

Can you help me, how to set limit for MACO considering toxicological data. I have set 10 ppm limit as general industrial practices. This i verified by MACO based on th daily dose criteria.
Pl. guide me.

[quote=PYB]Dear Sir,

Can you help me, how to set limit for MACO considering toxicological data. I have set 10 ppm limit as general industrial practices. This i verified by MACO based on th daily dose criteria.
Pl. guide me.[/quote]

Dear…

For MACO calculation most preferable approaches are 1. Based on Therapeutic daily dose 2. As per general limit criteria. Based on the mentioned approaches, we has been considering the lowest carryover from both approaches and limit shall be assigned 10 ppm or MACO value which ever is lower for total equipment train.

Generally toxicology data calculations performing when ever TDD is not available for the product [Generally such cases appeared for intermediates]. The calculations are as follows:

In cases in which a therapeutic dose is not known for intermediates/finished products toxicity data may be used for calculating MACO.
Calculate the NOEL number (No Observable Effect Level) according to the following equation and use the result for the establishment of MACO.

LD50 (g/kg) x 70 (kg a person)
NOEL = -----------------------------------------
2000
From the NOEL number a MACO can then be calculated according to:

                 NOEL x MBS

MACO = ------------------------------
SF x TDDnext

MACO: Maximum Allowable Carryover
NOEL No Observed Effect Level
LD50: Lethal Dose 50 in g/kg animal(mouse/rat etc.)
70 kg: 70 kg is the weight of an average adult
2000: 2000 is an empirical constant
TDDnext: Largest normal daily dose for the next product
MBS: Minimum batch size for the next product(s)
SF: Safety factor

Hi Prasad,
For MACO calculation, as one of the most preferred approach is to base on Therapeutic daily dose, what do you do when the changeover involves a respiratory product changing into an oral one, i.e. ug vs mg - this will result in a very tight MACO and probably cannot find an appropriate analytical method to match the calculated limit.

Regards
Amateur

[quote=amateur]Hi Prasad,
For MACO calculation, as one of the most preferred approach is to base on Therapeutic daily dose, what do you do when the changeover involves a respiratory product changing into an oral one, i.e. ug vs mg - this will result in a very tight MACO and probably cannot find an appropriate analytical method to match the calculated limit.

Regards
Amateur[/quote]

In such situations the analytical method shall be evaluated and develop the method to reduce LOD and LOQ limits up to attained MACO value.

Regards

[quote=jyopravel]In such situations the analytical method shall be evaluated and develop the method to reduce LOD and LOQ limits up to attained MACO value.

Regards[/quote]

Hi,
What do you do if you have done all you can and yet cannot find a method to match the calculated limit ? Do you spend all the time evaluating analytical method OR spending additional resource in equipment cleaning. Any perspective from the patient risk angle, i.e route of admininstration for Respiratory vs Oral

Regards

Thanks for sharing this knowledgeable information over here.

Basically, my post was to throw out this idea of calculating the MACO from the patient risk perspective for discussion, if we ever have to change over from a respiratory product to an oral one. Applying the MACO based on SF of 10x10x10 will invariably lead to an extremely tight limit (TDDugBS/SFTDDmg), and that makes the cleaning difficut or we may not even have a matching analytical method. Any views from this risk based perspective?

Regards
Amateur

Very informative.

Can you please Illustrate with an example by assuming two products.

Dear All,

Can anyone guide on how to calculate MACO / MAR when there is a changeover form two different API (A + B) to other API ©

i.e. 10 Equipments selected for product C, out of 10, 3 equipment was used for manufacturing of product A and 7 used for product B.

In this case how to calculate MACO.

very informative
thanks
sir

All,

i am using the PDA mACO calculation but am not sure if i have used the correct values. coudl one of you cast a critical eye over my calculations below and see what you think:

Our product is opthalmic and i have used the concentration of active in one drop (0.014mg) as my minimum therapetic dose, our smallest batch size of B is 200L so i have used 200000ml, safety factor of 1000 and for the max daily dose of product B i have used 40 drops per day with a drop size of 45ul so a total volume of 1.8ml. total surface area of filling machine is 7455cm2

0.014 x 200000 X 0.001 = 1.555mg (is this then my rinse sample acceptance criteria??)
1.8

4136.11 X 25cm2 = 0.0052mg/swab
7455

the 10ppm calculation comes out as 6.7mg per swab so significatnly higher.

we have been told in a recent audit also that we must use the tox data for future cleaning validaiotn calculations but our active ingredients are non toxic and have high Ld50 values so the therapetic dose criteria will also be so much lower.
i have calculated MAC using LD50 as follows:
NOEL:
1489 mg/kg = 0.7445 mg/kg/day
20000

ADI:0.7445 X 50 X 0.001 = 0.037225mg / day

MAC: 0.037225 X 200000 = 4136.111mg
1.8
4136.11 X 25cm2 = 13.869mg/swab
7455

In summary
10ppm - 6.70mg/swab
MACO therapeutic dose: 0.005mg/swab
Maco based on ADI/MACO: 13.869mg/swab

please advise

All,

i am using the PDA mACO calculation but am not sure if i have used the correct values. coudl one of you cast a critical eye over my calculations below and see what you think:

Our product is opthalmic and i have used the concentration of active in one drop (0.014mg) as my minimum therapetic dose, our smallest batch size of B is 200L so i have used 200000ml, safety factor of 1000 and for the max daily dose of product B i have used 40 drops per day with a drop size of 45ul so a total volume of 1.8ml. total surface area of filling machine is 7455cm2

0.014 x 200000 X 0.001 = 1.555mg (is this then my rinse sample acceptance criteria??)
1.8

4136.11 X 25cm2 = 0.0052mg/swab
7455

the 10ppm calculation comes out as 6.7mg per swab so significatnly higher.

we have been told in a recent audit also that we must use the tox data for future cleaning validaiotn calculations but our active ingredients are non toxic and have high Ld50 values so the therapetic dose criteria will also be so much lower.
i have calculated MAC using LD50 as follows:
NOEL:
1489 mg/kg = 0.7445 mg/kg/day
20000

ADI:0.7445 X 50 X 0.001 = 0.037225mg / day

MAC: 0.037225 X 200000 = 4136.111mg
1.8
4136.11 X 25cm2 = 13.869mg/swab
7455

In summary
10ppm - 6.70mg/swab
MACO therapeutic dose: 0.005mg/swab
Maco based on ADI/MACO: 13.869mg/swab

please advise

[quote=gokeeffe]
Cleaning validation is a process to ensure that equipment cleaning procedures are removing residues to predetermined levels of acceptability. Although “equipment cleaning” is part of current Good Manufacturing Practice requirements the term “cleaning validation” was not popular until late 1980s. The need for a systematic approach to proving the effectiveness of all the cleaning procedures was achieved in 1993 with a revised Food and Drug Administration Inspection Guide on Cleaning Validation.

Pharmaceutical products and active pharmaceutical ingredients (APIs) can be contaminated by other pharmaceutical products, by cleaning agents, micro-organisms or by other material (e.g. air-borne particles, dust, lubricants). Further sources of contamination might be raw materials, intermediates, auxiliaries, etc.

In many cases, the same equipment may be used for processing different products. To avoid contamination of the following product, adequate cleaning procedures are essential.

High risk products, such as penicillins, are a major concern in the cleaning validation field. Sensitive sampling methods require development and must be applicable to each specific piece of equipment used. Due to the possibility of inter-product contamination, highly sensitive analytical methods such as Liquid Chromatography are required for trace level analysis.

These detection methods should be specific for the target analyte, sensitive for trace and ultra-trace analysis, and be sufficiently able to separate and quantify the target analyte from potential interference. Currently there are two sampling methods in use.

The direct method, incorporating a swabbing material, is favoured over the indirect rinsate analysis approach. However caution must be taken when choosing the correct swab, as factors such as recovery, background contribution and particle generation can hinder residue determinations. Therefore each should be evaluated independently and an overall correction factor applied to the swab. Swab recoveries may be determined using spiking studies incorporating coupons of equipment surfaces.
Rinsate analysis is a useful sampling tool for equipment such as blenders and reaction vessels. The theory is that by analysing an aliquot of rinse water, the total quantity of analyte residue can be estimated. This method however assumes that the residue is uniformly removed from the equipment and also presumes that if the rinsate is clean then the equipment is clean.

Probably the most important aspect of cleaning validation programmes is establishing predetermined levels of acceptability. In order to establish Acceptable Residue Limits (ARL), various product and equipment attributes are evaluated. This leads to wide variations in ARL values between different product trains and different manufacturing facilities. The most important aspect is therefore proving that the ARL values determined in the cleaning validation programme have been established using a sound scientific rationale.[/quote]

All,

i am using the PDA mACO calculation but am not sure if i have used the correct values. coudl one of you cast a critical eye over my calculations below and see what you think:

Our product is opthalmic and i have used the concentration of active in one drop (0.014mg) as my minimum therapetic dose, our smallest batch size of B is 200L so i have used 200000ml, safety factor of 1000 and for the max daily dose of product B i have used 40 drops per day with a drop size of 45ul so a total volume of 1.8ml. total surface area of filling machine is 7455cm2

0.014 x 200000 X 0.001 = 1.555mg (is this then my rinse sample acceptance criteria??)
1.8

4136.11 X 25cm2 = 0.0052mg/swab
7455

the 10ppm calculation comes out as 6.7mg per swab so significatnly higher.

we have been told in a recent audit also that we must use the tox data for future cleaning validaiotn calculations but our active ingredients are non toxic and have high Ld50 values so the therapetic dose criteria will also be so much lower.
i have calculated MAC using LD50 as follows:
NOEL:
1489 mg/kg = 0.7445 mg/kg/day
20000

ADI:0.7445 X 50 X 0.001 = 0.037225mg / day

MAC: 0.037225 X 200000 = 4136.111mg
1.8
4136.11 X 25cm2 = 13.869mg/swab
7455

In summary
10ppm - 6.70mg/swab
MACO therapeutic dose: 0.005mg/swab
Maco based on ADI/MACO: 13.869mg/swab

please advise

Hi, our company also produces eye drops and now I am performing cleaning validation.I think that we have to take as an acceptance lower limit.

I see that this is an old thread, but I do sincerely hope that someone can benefit from what I am going to share. I have been working on remediation projects from the aftermath of people trying to set acceptance criteria based on the MAC or MACO calculations. I emplore upon you all to please not do this. While you can set to this, generally it is best to keep MAC/MACO in you back pocket as an investigational tool to justify why it is Okay to release if you happen to have a failure in your cleaning. The best option is to establish, through robust development work, that you are removing all of the soils. If you can demonstrate that you have established your cleaning process based on sound scientific understanding of you process soils and what it takes to remove them, then you can justify that you only need to test for TOC and conductivity during the rinse to demonstrate removal of the cleaning agents. The danger in having a spefic method that looks only for the specific soil is that other things will not be detected. By using conductivity and TOC, we can detect when there are other carbon elements that are not seen with specific methods. This is the current understanding that the FDA has from what they have learned over the years and is a very defendable position. The point a lot of people make is trying to reinvent the wheel when that requires much more effort than just following what the agancies are familiar with. I have not seen one successful defence of these other methods since they are very easy to poke holes in during an audit. Let’s face the reality of this whole situation and understand it is not what you think you can create, but what you can sucessfully convince the agencies of that keeps people in business.

Dear sir,
Informative article.

I do understand that the use of SS plate to perform the recovery study in cleaning validation is replicate the manufacturing equipment and is quite common to have a SS equipment for large scale production.But suppose my formulation is performed in a vessel other than Stainless steel (SS), then how do i proceed with the selection of plate to perform recovery?