EMA - Nitrosamines 19 July 2024
EMA - Nitrosamines 19 July 2024
EMA - Nitrosamines 19 July 2024
EMA/409815/2020 Rev.21
© European Medicines Agency, 2024. Reproduction is authorised provided the source is acknowledged.
Table of contents
Revision History ............................................................................................ 3
Introduction ................................................................................................. 4
1. Should the risk of presence of nitrosamines be considered for all human medicinal
products? ..................................................................................................... 5
2. What is the ‘call for review’? ....................................................................... 6
3. For the ‘call for review’ for chemically synthesised and biological medicinal
products, when and how should MAHs report steps 1 and 2 to competent authorities?
4. What are the currently identified risk factors for presence of nitrosamines? ....... 9
5. What to do if after submission of step 1 and /or step 2 responses, new information
(e.g. related to new potential risk factors or root causes) is identified? ............... 11
6. What factors should be considered in prioritising the risk evaluation? .............. 12
7. How should the risk evaluation be performed? ............................................. 12
8. How should confirmatory tests be conducted by MAHs and manufacturers?...... 13
(Updated) .................................................................................................... 7
9. What are the requirements of the analytical method(s)? (Updated) ................ 15
10. Which limits apply for nitrosamines in medicinal products? (Updated) ........... 15
11. What should I do if a nitrosamine is detected in my medicinal product? ......... 20
12. Which are the measures to mitigate the risk of presence of nitrosamines? ..... 20
13. Which changes would be required for Marketing Authorisations? ................... 21
14. What is the approach for new and ongoing marketing authorisation applications
(MAA)? ........................................................................................21 (Updated)
15. When should a test for nitrosamines be included in the MA dossier? 23 (Updated)
16. What are the responsibilities of MAHs for APIs with CEPs or ASMFs?23 (Updated)
17. How does the lessons learnt exercise from presence of nitrosamines in sartans
relate to the Article 5(3) Referral Outcome? .................................................... 24
18. What about regulatory requirements in other regions? ................................ 24
19. What is the approach for line extensions and variations applications not linked to
changes required as part of article 5(3) recommendation? ................................ 24
20. What are the regulatory steps taken by authorities following the identification of
an N-nitrosamine exceeding the AI? .............................................................. 25
21. What is the approach to control the presence of nitrosamines until a substance
specific AI is established? ............................................................................. 25
22. What is the approach to control presence of N-nitrosamine exceeding the AI
during CAPA implementation? ....................................................................... 25
Annex 1: Decision tree with control options for products containing multiple N-
nitrosamines: ............................................................................................. 28
0 Replace obsolete Q&A published in 2019 to support the initial “call for 03rd August 2020
review” with a new version reflecting the main principles agreed as
part of the Article 5(3) referral which concluded in July 2020.
1 Update to Q&A 3 in order to clarify products in scope of the call for 29th January 2021
review.
Update to Q&A 4 in order to add the link to the outcome of the referral
under article 3 of Directive 2001/83/EC for ranitidine.
2 Update to Q&A 3 on indicating testing timeline at the time of step 1 24th February 2021
“risk identified” reporting.
3 Update to Q&A 3 on the approach for non-marketed medicines. 15th April 2021
New Q&A 19 on the requirements for line extensions and variation
applications.
4 Update to Q&A 3 on combining step 2 response for multiple products 18th May 2021
from the same MAH.
4* Updates to Q&A 3 on when to perform step 2 confirmatory testing in 29th June 2021
order to meet the established deadline for step 3.
Update and Q&A 10 to add an AI for NMOR.
5 Update to Q&A 10 to add an AI for NNV. 21st September 2021
6 Guidance on confirmatory testing requirements for marketed (Q&A 8) 14th October 2021
and on-going applications (Q&A 14) to include cases where a potential
nitrosamine impurity cannot be synthesised, and when a product is
available in multiple strengths of the same dosage form.
7 Inclusion of additional guidance on control strategies for products 31st January 2022
containing more than one nitrosamine impurity including examples
(Q&A 10) and a decision tree (Annex I).
8 Update to guidance on root causes and risk factors for nitrosamine 24th March 2022
contamination (Q&A 4) and on policy for confirmatory testing (Q&A 8)
and dossier requirements (Q&A 15) to allow testing of intermediates,
raw materials or API under certain circumstances.
9 New Q&A 20 providing clarifications on what are the regulatory steps 20th May 2022
for dealing with scenario A cases and update Q&A10 with new AIs
(N-nitrosomethylphenidate, N-nitrosopiperidine, N-nitrosorasagilene,
7-Nitroso-3-(trifluoromethyl)-5,6,7,8-tetrahydro[1,2,4]triazolo-[4,3-
a]pyrazine, N-nitroso-1,2,3,6-tetrahydropyridine, N-
nitrosonortriptyline, N-methyl-N-nitrosophenethylamine) and
guidance on use of Ames test.
10 Update to Q&A 5 to provide clarifications on the expectation for MAHs 23rd June 2022
to continue to re-visit risk evaluations when new information becomes
available with specific reference to API-nitrosamine risk. Update to
Q&A 10 to include newly adopted AI for N-nitrosodabigatran and to
indicate APIs where related nitrosamines have been identified.
Clarification of how to set limits for products containing salt, hydrate
or solvate forms of the API. Update to Q&A 14 to reference the new
risk evaluation template for use in marketing authorisation
applications.
11 Update to Q&A 3 on submission of amended step 1 response and 29th July 2022
extension of Step 3 deadline for chemical medicines.
12 Update of Q&A 10 to add nitrosoduloxetine and introduction of Q&A 21 10th October 2022
on approach to control presence of nitrosamine while the AI is being
established.
13 Update of Q&A 10 to add N-nitrosofluoxetine, N-nitrosoparoxetine, N- 5th December 2022
nitrosodiphenylamine, N-nitroso-mefenamic acid, N-nitrosopyrrolidine
and N-nitrosodiethanolamine.
14 Introduction of Q&A 22 on approach to control presence of N- 22nd December 2022
nitrosamine exceeding the AI while CAPAs are being implemented.
Update of Q&A 20 to consider the possibility of an interim limit based
on the LTL approach during CAPA implementation. Update of Q&A 21
Introduction
The assessment report of the CHMP’s Article 5(3) of Regulation (EC) No 726/2004 opinion on nitrosamine
impurities in human medicinal products provides general guidance and recommendations on mitigating
and preventing the presence of nitrosamines in human medicinal products. In this context all
MAHs/Applicants of human medicinal products should work with the manufacturers of their Active
Pharmaceutical Ingredients (APIs) and finished products (FPs) in order to ensure that the presence of
nitrosamine impurities in their medicinal products is mitigated as much as possible and controlled at or
below a limit defined based on ICH M7(R2) principles for substances of the “cohort of concern” reflected
in this guideline and calculated considering a lifetime daily exposure and kept as low as possible and that
appropriate risk mitigating measures are taken.
The published CHMP Article 5(3) opinion, supplemented by the current Question and Answer document
on its implementation, will replace the previous letter entitled ‘Information on nitrosamines for marketing
authorisation holders’ (EMA/189634/2019, published on 19 September 2019).
The terms “nitrosamine” and “N-nitrosamine” are used interchangeably within this Q&A and related
documents and should both be understood to refer to the following structure:
For details on the approach required, please refer to Q&A 10 on the limits for nitrosamines and Q&A 12
on the measures to mitigate the risk of presence of nitrosamines.
MAHs/Applicants are reminded of their obligations to ensure that, in accordance with Article 23 and
Annex I of Directive 2001/83/EC and Article 16 of Regulation (EC) No 726/2004, their medicinal products
are manufactured and controlled by means of processes and methods in compliance with the latest state
of scientific and technical progress.
• design their manufacturing processes and controls to prevent if possible or mitigate as much as
possible the presence of N-nitrosamines in their API and FP(s);
• assess the risk of presence nitrosamine impurities in their API(s) and FP(s) and introduce any
resultant changes to the dossier as needed (e.g. changes to their manufacturing processes);
1
https://www.ema.europa.eu/en/documents/referral/nitrosamines-emea-h-a53-1490-information-nitrosamines-marketing-
authorisation-holders_en.pdf
2
https://www.ema.europa.eu/en/documents/referral/nitrosamines-emea-h-a53-1490-questions-answers-information-
nitrosamines-marketing-authorisation_en.pdf
Compliance of the MAHs/Applicants with the above-mentioned obligations is subject to regular controls
by competent authorities including during GMP inspections.
While the Article 5.3. recommendations on controlling nitrosamine impurities apply to all human
medicinal products, the call for review applies only to human medicines containing chemically
synthesised APIs or biological APIs, as further explained in Q&A 2 below.
Following the conclusion of the review under Article 5(3), the CHMP considered that there is also a risk
of presence of nitrosamines in biological medicinal products, in particular for the biological medicines
with the following risk factors:
• biologicals containing chemically synthesised fragments, where risk factors similar to chemically
synthesised active substances are present;
• biologicals packaged in certain primary packaging material, such as blister packs containing
nitrocellulose.
For the above reasons the current call for review has been extended to cover also all biological
medicinal products for human use. For further reference on what is considered to be a biological
medicinal product for the purpose of this exercise, please consult the CMDh Questions & Answers on
Biologicals.
• Step 1: MAHs to perform a risk evaluation to identify if APIs and/or FPs could be at risk of presence
of nitrosamine;
• Step 2: if a risk is identified, MAHs to proceed with confirmatory testing in order to confirm or refute
the presence of nitrosamines. MAHs should report outcomes as soon as possible;
• Step 3: if the presence of nitrosamine(s) is confirmed, MAHs should implement effective risk
mitigating measures through submission of variation.
Please refer to Q&A 3 for further details on the ‘call for review’ including the timelines for chemicals and
the timelines for biologicals.
For the specific case of sartans with a tetrazole ring that have been subject to a review under Article 31
of Directive 2001/83/EC, further guidance will be published soon.
Products that have been approved after 26 September 2019 but for which a risk evaluation was not
assessed within the MAA procedure should comply with the call for review deadlines, if not already done
so.
For product containing chemically synthesised APIs, the step 1 risk evaluation should be concluded and
reported at the latest by 31st March 2021.
For product containing biological APIs, step 1 risk evaluation should be concluded and reported at the
latest by 01st July 2021.
The risk assessment has to be performed for all products for which a potential risk has been identified
in step 1, irrespective of the marketing status of the product or whether any registered manufacturers
are actively used in supply. However, it is recognised that step 2 may not be possible for medicines that
are not marketed, including the case of manufacturers not actively used in supply, since there may be
no finished product batches available for confirmatory testing. In these cases, i.e. where no batches of
finished products are available, it would be acceptable to submit a written commitment that step 2
confirmatory testing will be conducted once finished product has been manufactured and/or the product
is launched. The outcome of step 2 testing as well as any necessary variation(s) as part of step 3 will
therefore need to be submitted and approved before the product can be placed on the market or the
manufacturer can be actively used in supply, even if this is after the step 2 and 3 deadlines.
MAHs’/Applicants’ compliance with the above-mentioned obligations is subject to regular controls by
competent authorities including during inspections.
All MAHs should inform the concerned Competent Authorities of the outcome of their risk evaluation
(step 1) using the dedicated templates.
If a risk has been identified, the expected timeline for the testing activities should also be provided as
foreseen in the dedicated template. No additional documentation is required at this stage. However, the
risk review should be adequately documented, and related documentation should be made available
upon request.
Step 2 should be started as soon as a risk is identified in API and/or FP and in accordance with product
prioritisation (see Q&A 6).
If a risk has been identified for the API, the MAH is advised to report this outcome by using step 1
response template and to proceed directly to step 2 confirmatory testing of the FP. If no risk has been
identified in the API, the MAH is advised to proceed with the risk evaluation of the FP and to present the
result of Step 1 when a final conclusion has been reached on both the API and the FP. MAHs should
inform the concerned Competent Authorities of the outcome of their risk evaluation (step 1) even if no
risk has been identified in the API or FP.
It is acceptable for the submission of the outcome of step 1 to submit one email notification grouping
products with identical outcome under the following provisions:
• For those Member States that have a dedicated portal, the MAH should submit the notification via
this portal;
In specific cases it may be possible to correct a former step 1 outcome from “risk” to “no risk” by using
the “Step 2 no nitrosamine detected response template”. This template now contains a tick box for such
cases. The possibility to amend the step 1 outcome may only be used in those cases where data was
missing at the March 2020 deadline and is now available.
Submission of step 2 outcome
The step 2 confirmatory testing should be conducted in accordance with product prioritisation (see
Q&A 6).
For nitrosamine impurities that are classified as non-mutagenic in Appendix 1 based on in-vivo
mutagenicity studies, the submission of step 2 confirmatory testing is not required, and these impurities
should be controlled according to ICH Q3A(R2) and ICH Q3B(R2) guidelines. For all other nitrosamine
impurities the submission of step 2 confirmatory testing is required using the dedicated response
templates.
For product containing chemically synthesised APIs, confirmatory testing activities at Step 2 are
expected to be finalised at the latest by 26th September 2022. MAHs should refrain from submitting
incomplete step 2 outcomes.
The deadline for the submission of any changes required to Marketing Authorisations (Step 3, see
Q&A 13) is by 1st October 2023.
For product containing biological APIs, confirmatory testing activities at Step 2 and submission of any
changes required to Marketing Authorisations (Step 3, see Q&A 13), are expected to be finalised at the
latest by 1st July 2023.
In order to meet the above deadlines for submission of any changes required to Marketing Authorisations
at Step 3 for products containing chemically synthesised or biological APIs, it would be expected that
confirmatory testing activities at Step 2 are finalised in advance of these deadlines.
MAHs should forthwith inform the competent authorities if tests confirm the presence of nitrosamine,
irrespective of the amount detected and by utilising the dedicated reporting templates. The immediate
risk to patients should be assessed based on the limits defined in Q&A 10 and appropriate action proposed
to avoid or minimise the exposure of patients to nitrosamines.
For the submission of the outcome of step 2 confirmatory testing several products can be combined
when the outcome is “no nitrosamines detected”. When the outcome is “nitrosamines detected” all
strengths and pharmaceutical forms of one marketing authorisation can be combined in one response
template when the supporting documentation is completely identical for all products concerned; if not
the response has to be submitted separately.
In case one or more nitrosamines are identified that exceed the limit defined in Q&A 10, the following
supportive documentation is required at the time of reporting:
• interim investigation report including (preliminary) root cause, risk mitigating plan and benefit/risk
assessment.
For their responses, MAHs are required to use dedicated templates and contact points as outlined on the
EMA and CMDh websites.
MAH(s) are reminded of their responsibilities to ensure the quality, safety and efficacy of their medicines
and to adhere to the Nitrosamines guidance outlined by the EU Network. MAHs and Manufacturers should
work together and take precautionary measures to mitigate the risk of presence of nitrosamines during
the manufacturing and storage of all authorised medicinal products and throughout the lifecycle of the
product if any changes are made.
Authorities in the EU will continue take all necessary measures to protect patients and ensure that
medicines in the EU meet the required quality standards. The Authorities in the EU will also continue to
collaborate with international partners on Nitrosamine to reflect scientific advances.
1. Use of nitrite salts and esters (e.g. NaNO2, alkyl nitrites), or other nitrosating agents (e.g. nitroso
halides, nitrosonium salts, nitrogen oxides, nitro alkanes, halogenated nitro alkanes, Fremy’s salt,
nitroso sulfonamides),3,4 in the presence of secondary or tertiary amines within the same or
different steps of the manufacturing process. Sources for secondary or tertiary amines can also
be starting materials, intermediates, reagents, solvents (e.g. DMF, DMAc and NMP) and catalysts,
which contain amine functionality, amine impurities (e.g. quaternary ammonium salts) or which
are susceptible to degradation to reveal amines.
3
Lessons learnt from presence of N-nitrosamine impurities in sartan medicines EMA/526934/2019.
4
Org. Process Res. Dev. 2020, 24 (9), 1558–1585
5
Chem. Rev. 2016, 116, 422−518
4. Oxidation of hydrazines, hydrazides and hydrazones by hypochlorite, air, oxygen, ozone and
peroxides in the manufacturing process or during storage.4
5. Use of contaminated raw, recovered or recycled materials (e.g. solvents, reagents and catalysts)
in the API manufacturing process.
6. Use of contaminated starting materials and intermediates supplied by vendors who use processes
or raw materials which may contain residual nitrosamines or nitrosating agents.
9. Degradation processes of active substances, including those induced by inherent reactivity (e.g.
presence of nitro-alkyl, oxime, or other functionality311,4) or by the presence of an exogenous
nitrosating agent. This could potentially occur during both active substance and finished product
manufacturing processes or during storage and could be influenced by crystal structure, crystal
habit and storage conditions (temperature, humidity etc.). For more details, refer to page 6 of
Referral under Article 31 of Directive 2001/83/EC for ranitidine and published literature.11,12
10. Oxidation of hydrazine or other amine-containing functional groups present in active substances
or their impurities/degradants (e.g. from hydrazones and hydrazides), either in active substance
manufacturing processes or during storage.4 This root cause has also been observed during
11. Use of certain packaging materials. Relevant nitrosamine contamination has been observed in
primary packaging of finished products in blister with lidding foil containing nitrocellulose. During
the blister heat-sealing process, nitrogen oxides can be generated thermally from nitrocellulose.
Under these conditions, nitrosamines have been shown to form from low molecular weight amines
present either in printing ink or in the finished product and to transfer to the product and/or to
the cavity via evaporation and condensation.
12. Reaction of amines leaching from quaternary ammonium anion exchange resins (e.g. used for
purification steps) with nitrosating agents present in the liquid phase. A recent example of this
was in the production of water for injections where residual chloramine used to disinfect incoming
water reacted with dimethylamine leaching from the anion exchange resin used in the
demineralisation step to form NDMA. In addition, disinfection procedures such as e.g. chlorination,
chloro-amination and ozonisation can lead to significant N-nitrosamine generation as by-products
in case vulnerable amines are present.6,7,8,9 Given the source of contamination, risk is related to
the concentration of the reactive agent(s) and thus to the volume of water in or used to dilute a
particular product. The same risks could be associated with active substances or finished products
manufactured using water purified using similar resins.
13. Cross-contamination due to different processes being run successively on the same
manufacturing line.
14. Carry-over of impurities between process steps due to operator-related errors or insufficiently
detailed batch records such as inadequate phase separations during work-up procedures.
15. Use of contaminated recovered or recycled materials (e.g. solvents, reagents and catalysts)
where the recovery is outsourced to third parties who are not aware of the content of the materials
they are processing. Recovery processes carried out in non-dedicated equipment should also be
considered.
In particular, MAHs should note the risk of formation of nitrosamine impurities from active substances
(or their related impurities) containing a vulnerable amine during finished product formulation and/or
storage due to the presence of traces of nitrites. This has been recently elaborated as a risk factor to
Q&A 4 (bullet 8) based on understanding gained during the call for review. MAHs that did not take into
account this risk as part of step 1 response for their products containing active substances with
vulnerable amines should reconsider their original step 1 risk evaluations in light of this new information
and proceed to step 2 confirmatory testing as appropriate (see also Appendix 1).
The same approach should be followed for medicinal products granted a positive opinion and marketing
authorisation during the call for review.
In order to undertake the analysis of the identified medicinal products at risk, MAHs can also use tools
such as Failure Mode Effects Analysis (FMEA) and Failure Mode, Effects and Criticality Analysis (FMECA)
as outlined in the ICH Q9 guideline on quality risk management.
Manufacturers of active substances and FP and their raw material suppliers should provide
MAHs/applicants with all information necessary for a comprehensive risk evaluation. If the risk of
nitrosamine impurity formation was assessed during the development phase of the API/FP manufacturing
processes, the information from this assessment can be used to support the risk evaluation.
MAHs/Applicants and manufacturers should consider as part of the risk evaluation all potential sources
of contamination or formation of nitrosamine, notably the root causes listed under Q&A 4.
As MAHs/Applicants and manufacturers for products containing biological APIs should consider the
following aspects that may increase the risks of nitrosamine presence in their products:
• biologicals containing chemically synthesised fragments, where risk factors similar to chemically
synthesised active substances are present;
• biologicals packaged in certain primary packaging material, such as blister packs containing
nitrocellulose.
If, after completion of the risk evaluation, a risk is identified in the API and/or the FP, MAHs/applicants
must notify the competent authorities of the identified risk, proceed without further delay with
confirmatory tests (see Q&A 8) and introduce any necessary changes to the dossier.
All MAHs should inform the concerned Competent Authorities of the outcome of their risk evaluation
(step 1) even if a risk has not been identified, please see Q&A 3 for further details.
However, some root causes may only be linked to the API manufacturing process (see Q&A 4). In these
cases, testing of the API or intermediates upstream of the active substance could be used as a surrogate
for testing the finished product, provided that the risk assessment performed on the FP concluded no
additional risk factors for formation of nitrosamine impurities in the finished product (see Q&A 4, risk
factors related to the finished product). If testing is carried out on an intermediate, then there should
also be no risk factors associated with subsequent steps in the API manufacturing process or the finished
product. The confirmatory testing strategy is the responsibility of the MAH and should be justified based
on the risk assessment for the finished product and documented in the MAH’s pharmaceutical quality
system. It should be clearly justified why testing of the active substance or intermediate is appropriate
and why further risk of nitrosamine formation in the finished product or subsequent API manufacturing
steps can be excluded. If nitrosamines are detected, then an appropriate control strategy should be
implemented in the dossier.
In any case, if the control point of nitrosamines is not in the finished product, the responsibility for quality
lies with the MAH.
The number of batches to be tested should be commensurate with the risk. MAHs and manufacturers
should test a representative number of batches of FP and the relevant starting materials, intermediates,
API or raw materials as applicable. If the source of risk has been identified and is well understood (e.g.
by spike and purge studies) such that impurity levels are expected to be consistent from batch to batch,
testing should be conducted on 10% of annual batches, or 3 per year, whichever is highest. This includes
testing not only of newly produced batches but also retained samples of batches still within expiry date.
If fewer than 3 batches are manufactured annually, then all batches should be tested.
If multiple manufacturers, manufacturing processes and/or sources of at-risk raw materials are used,
(or were used historically for batches still within expiry date), then testing of additional batches would
be necessary to cover these risk factors.
If a product is available in multiple strengths of the same dosage form with the same risk factors
applicable to each, then testing could be rationalised by testing only the worst-case scenario strength.
During development of an analytical method, a reference standard of the relevant nitrosamine impurity
is generally needed. If, despite extensive efforts, it becomes apparent that the relevant nitrosamine
impurity cannot be synthesised, then this could be an indication that the nitrosamine either does not
exist or that there is no risk of it being formed. In such cases, it may not be necessary to conduct
confirmatory testing. This should be justified thoroughly on a case-by-case basis according to appropriate
scientific principles. The justification could include relevant literature, information on structural/stereo-
electronic features and reactivity of the parent amine, stability of the nitrosamine and experimental data
to illustrate the efforts made to synthesise and to analyse the impurity. The justification should be
documented in the risk assessment in the MAH's pharmaceutical quality system.
For products where nitrosamine impurities can be controlled according to ICH Q3A/B principles, see Q&A
10, confirmatory testing is generally not needed if the risk can be sufficiently mitigated based on scientific
considerations that demonstrate that the relevant ICH Q3A/B thresholds will not be exceeded. In such
cases, the justification should be documented in the risk assessment in the MAH's pharmaceutical quality
system. For products with only advanced cancer indications as defined within the scope of ICH S9, and
where the above principle has been applied to mitigate the potential risk of nitrosamine impurities, the
approach should be revised should the indication ever move beyond the relevant ICH S9 setting. In these
instances, confirmatory testing would generally be required considering the resultant lower applicable
limits when ICH Q3A/B ceases to apply.
Methods for determination of various nitrosamines in sartans with a tetrazole ring, metformin and
ranitidine have already been developed by the Official Medicines Control Laboratories and are available
for reference on the European Directorate for the Quality of Medicines & HealthCare (EDQM) website.
These may serve as a starting point for the development and validation of analytical methods for testing
other APIs/FPs.
Appropriately sensitive analytical methods for determination of specific nitrosamines in other medicinal
products should be developed, see Q&A 9 and validated accordingly before testing.
Given the trace levels of nitrosamines to be measured, the following technical aspects should be
considered when developing analytical methods:
• Interference caused by presence of trace amounts of nitrosamines in testing materials utilised (e.g.
water, airborne sources, plastics products and rubber/elastomeric products);
• Contamination during sample preparation (avoiding cross contaminations from gloves, membranes,
solvents etc.) which could lead to false positive results;
• Use of accurate mass techniques are required (MS/MS or high-resolution accurate mass systems) in
order to overcome interference in the identification of the specific peak of a certain nitrosamine (e.g.
false positives have been observed from DMF co-eluting with NDMA).
As a result of the above considerations, control experiments should be conducted such as analysing
samples by orthogonal analytical methods.
Further details in relation to analytical methodology can be found on EDQM website and in the CHMP
assessment report of the CHMP’s Article 5(3) opinion on nitrosamine impurities in human medicinal
products.
• The limit of quantification (LoQ) provides the minimum level at which an analyte can be quantified
with acceptable accuracy and precision and should thus be used for impurity testing and decision-
making;
• If quantitative testing is performed as a routine control, the LoQ should be ≤ of the acceptable limit
based on the relevant acceptable intake (AI) for the respective nitrosamine impurity;
• If quantitative testing is performed to justify skip testing, the LoQ of the analytical procedure
employed should be ≤ 30% of the acceptable limit based on the AI;
• If quantitative testing is performed to justify omission of specification, the LoQ of the analytical
method employed should be ≤ 10% of the acceptable limit based on the AI;
• Exceptions are anticipated for medicinal products used at high daily doses (AI may be below technical
feasibility of the method), or in case more than one nitrosamine is anticipated or identified in a given
medicinal product.
Different analytical methods may be used for determination of multiple nitrosamines. If the same
analytical method is used for multiple nitrosamines, the selectivity of the method should be demonstrated
for each nitrosamine.
For products where nitrosamine impurities can be controlled according to principles of ICH Q3A/B, see
Q&A 10, where tested the LoQ for the analytical procedure should be at or below the applicable reporting
threshold for the respective drug substance and/or drug product.
The ‘less than lifetime’ (LTL) approach should not be applied in calculating the limits as described above
but can only be considered after consultation with competent authorities as a temporary measure until
further measures can be implemented to reduce the contaminant at or below the limits defined above.
For products intended for advanced cancer only as defined in the scope of the ICH S9 guideline, N-
nitrosamine impurities should be controlled according to ICH Q3A(R2) and ICH Q3B(R2) guidelines, as
specified in the Q&A document to ICH S9 guideline. In addition, limits according to ICH Q3A(R2) and
The same risk approach is applicable to all routes of administration. Corrections to limits are generally
not acceptable unless route-specific differences are justified by data.
A. If N-nitrosamines are identified with sufficient substance specific animal carcinogenicity data, the
TD50 should be calculated and used to derive a substance specific limit for lifetime exposure as
recommended in ICH M7(R2) guideline.
B. If N-nitrosamines are identified without sufficient substance specific data to derive a substance
specific limit for lifetime exposure as recommended in ICH M7(R2) guideline,
2. A negative result in an GLP-compliant enhanced Ames test (EAT, Appendix 3) allows control of
the N-nitrosamine at 1.5 µg/day. For substances testing positive, the AI should be established
using options 1 or 3. For reporting requirements see Q&A 3 above.
3. If a surrogate nitrosamine is available with sufficiently robust carcinogenicity data, the TD50
from the surrogate substance can serve as a point of departure for derivation of AI by SAR and
read across.
4. A negative result in a relevant well-conducted in vivo mutagenicity study can allow control of the
N-nitrosamine as a non-mutagenic impurity (NMI), i.e. according to ICH Q3A(R2) and ICH
Q3B(R2) limits, irrespective of the limit calculated through option 1, 2 or 3. For substances
testing positive, the AI should be established using options 1 or 3. For reporting requirements
see Q&A 3 above.
The risk approach is applicable to all routes of administration. Corrections to limits are generally not
acceptable unless data justify route-specific differences.
Appendix 1 lists the nitrosamines for which acceptable intakes have been established by the Non-clinical
Working Party. If the nitrosamine is not included in Appendix 1, MAH/MA applicants can also refer to a
CPCA category from another source e.g. CPCA categories published by other regulatory authorities, but
this will need confirmation to allow control of the substance at the level corresponding to that category.
All Ames assays initiated after August 2023 must comply with the requirements of the EAT protocol
(Appendix 3).
The conversion to a specification limit in ppm for a particular medicinal product is calculated by dividing
the respective AI in Appendix 1 (ng) by the maximum daily dose (mg) of a given product as reflected in
the SmPC.
The maximum daily dose is defined in line with the definition of the product strength in the Guideline on
the SmPC. Therefore, the limit in ppm should usually be expressed per active moiety (free base, free
For a control point in the API only, the limit should be expressed in general per drug substance (i.e.
relating to form of salt, hydrate, solvate etc. where relevant).
Calculation of limit when more than one nitrosamine is identified in the same product
Note: This section does not apply to scenarios where the nitrosamine impurities are intended to be
controlled according to ICH Q3A/B principles.
Please also refer to the decision tree in Annex 1 for further guidance.
For determining limits in the case of presence of more than one nitrosamine, two approaches are
considered acceptable in order not to exceed the acceptable risk level of 1:100,000 as outlined in ICH
M7(R2) guideline:
1. The total daily intake of all identified N-nitrosamines not to exceed the AI of the most potent N-
nitrosamine identified, or
2. Total risk level calculated for all identified N-nitrosamines not to exceed 1 in 100,000.
Specifications for individual N-nitrosamines should generally include an AI limit expressed in ppm or ppb.
The conversion to an AI limit in ppm/ppb for a particular medicinal product is calculated by dividing the
respective above AI (in ng/d) by the maximum daily dose (in mg) of a given product as reflected in the
SmPC. The calculation of the specification limit does not take into account the molecular weight of the
N-nitrosamine.
It is considered that the presence of one or more N-nitrosamines at <10% of their respective AI
constitutes a negligible toxicological risk, and as such, they do not need to be specified. N-Nitrosamines
present below 10% of their respective AI do not need to be factored into the calculation of limits for
individual or total N-nitrosamine(s).
However, the overall principle of the Article 5(3) referral should still be considered, notably that “the
presence of N-nitrosamines in human medicinal products shall be mitigated as much as possible.”
Therefore, manufacturers are encouraged to improve their processes, even if they result in only very
small amounts (<10% AI) of multiple nitrosamines, as processes and controls should be designed to
prevent if possible or mitigate as much as possible the presence of N-nitrosamines in APIs and FPs (see
Q&A 1).
For option 1, the AI limit for total N-nitrosamines should be set in ppm/ppb according to the most potent
N-nitrosamine present at ≥ 10% of its AI. The most potent nitrosamine is the one with the lowest AI
(see Appendix 1). Limits for individual N-nitrosamines can be defined but are not necessarily needed.
However, it should be clearly stated which N-nitrosamines are included in the calculation of total N-
nitrosamines.
For option 2, the limits for N-nitrosamines should ensure an overall risk of not more than 1 in
100,000. Different approaches can be employed to achieve this risk requirement:
Fixed approach: fixed AI limits (in ppm/ppb) are set for individual nitrosamines and no limit for total
N-nitrosamines is needed. The limit for each N-nitrosamine should be set at a percentage of its AI limit
such that the sum of the % AI limits for each specified nitrosamine does not exceed 100%.
Where Xi is the amount of each single N-nitrosamine i in ppm and AIi is the AI limit of each N-nitrosamine
i in ppm.
For each batch, to determine whether the limit for total N-nitrosamines is met, the amount of each N-
nitrosamine present (in ppm/ppb) should be converted to a percentage of its respective AI limit. The
sum of % AI limits of specified N-nitrosamines should not exceed 100%.
Example of control options and specifications for multiple nitrosamines in the same finished
product:
Example:
NDMA and NDEA are both detected at or above 10% of their respective AI) in a finished product with
maximum daily dose of 300 mg.
AI limit
• NDEA: 26.5 ng/day / 300 mg/day = 0.088 ppm or 88 ppb = most potent N-nitrosamine
• NDMA: 96.0 ng/day / 300 mg/day = 0.32 ppm or 320 ppb
Specification possibilities for different control options:
could be used in different situations dependent on relative amounts present, provided that the sum of the % AI limits
for each specified nitrosamine does not exceed 100%.
(50% of AI)
Total NA NMT 88 ppb 82 ppb Not needed - NMT 100% 62%
Mutagenic APIs are defined as substances having DNA-reactive properties as described in ICH M7.
The ICH M7(R2) guideline does not apply to drug substances and drug products intended for advanced
cancer indications as defined in the scope of ICH S9 (Ref. 4). Additionally, there may be some cases
where a drug substance intended for other indications is itself genotoxic at therapeutic concentrations
and may be expected to be associated with an increased cancer risk. Exposure to a mutagenic impurity
in these cases would not significantly add to the cancer risk of the drug substance. Therefore, impurities
could be controlled at acceptable levels for non-mutagenic impurities. Below it is explained in more detail
how this is applied to the control of nitrosamine impurities.
ii. The rules established for the control of nitrosamines as explained in the Article 5(3)
referral or elsewhere in the Q&A apply when mutagenicity/clastogenicity of API is
considered not to produce a significant risk for mutagenicity/clastogenicity at therapeutic
exposures.
i. The rules established for the control of nitrosamines as explained in the Article 5(3)
referral or elsewhere in the Q&A apply since aneugenicity of API is considered not to
produce a significant risk for carcinogenicity at therapeutic exposures
i. The rules established for the control of nitrosamines as explained in the Article 5(3)
referral or elsewhere in the Q&A apply
Higher limits may be set for nitrosamines in certain cases. However, it is expected that the Applicant/MAH
will ensure that the presence of nitrosamine impurities in their medicinal products is mitigated as much
as possible.
1Wherever it is quoted ”Control nitrosamine at or below ICH Q3A/B qualification threshold”, this implies
that control at the qualification threshold is justified from a safety perspective.
The MAH/Applicant should forthwith inform the competent authorities, irrespective of the amount
detected as described in Q&A 3 for medicinal products subject to the call for review.
The levels should be reported in ng and ppm, together with the corresponding calculations used to
describe the potential exposure to the detected nitrosamine based on the maximum daily dosage
recommended in the SmPC. If SmPCs differ between Member States, the calculations should be provided
for each different maximum exposure. Sufficient details should be provided to enable the calculations to
be reviewed and verified.
The calculated exposure(s) should then be compared to the limit defined in Q&A 10:
• If the limit is not exceeded for the detected nitrosamine or, in case of presence of multiple
nitrosamines, if the total risk remains below a theoretical lifetime excess risk of <1:100,000, the
MAH/Applicant shall control the nitrosamine(s) in the FP at or below this limit (see Q&A 10) and
should take measures to mitigate the risk of nitrosamine formation or contamination in the medicinal
product as much as possible (see Q&A 12).
• Where the limit defined in Q&A 10 for single or multiple nitrosamines is exceeded, the MAH/Applicant
should submit forthwith an (interim) investigation report including (preliminary) root cause, risk
mitigating plan and benefit/risk assessment. The competent authorities will then assess the impact
on the benefit/risk balance and the consequent need for any action to be taken.
Please refer to the Assessment report of the CHMP’s Article 5(3) opinion on nitrosamine impurities in
human medicinal products for further information.
Changes to the marketing authorisation related to measures to prevent or minimise the risk should be
introduced without delay and in accordance with the guideline on classification of variation (please refer
to Q&A 13).
If the presence of specific nitrosamine(s) in a medicinal product has already been reported to
the authorities by the MAH and is below the limit defined in Q&A 10 or a limit approved by the
authorities, there is no need for a further notification to the authorities.
MAHs shall design or adapt the manufacturing process of their medicinal products to prevent formation
of and contamination with nitrosamines whenever possible.
MAHs should implement a control strategy regarding N-nitrosamines, which should include current and
prospective measures to minimise the risk of generation of/contamination with nitrosamines (e.g. change
of manufacturing process, change of raw material quality, introduction of appropriate specifications and
MAHs shall also ensure that active substances and excipients used in their FPs are manufactured in
compliance with good manufacturing practices in line with Article 46(f) of Directive 2001/83/EC.
Please refer to the Assessment report of the CHMP’s Article 5(3) opinion on nitrosamine impurities in
human medicinal products for further information.
When nitrosamine(s) is (are) identified, the corresponding limit(s) as defined in Q&A 10 should be
introduced in the specifications of the FP. Please refer to Q&A 15 for information on the test modalities.
The application for a variation should contain information on amendments to the marketing authorisation
– i.e. in module 3 (3.2.S and 3.2.P), the active substance master files (ASMF) or the Certificate of
Suitability to the monographs of the European Pharmacopoeia (CEP) that is necessary to control
nitrosamine impurities in the active substance and/or FP. Variations should be submitted according to
the existing variations classification guideline: EUR-Lex - 52013XC0802(04) - EN - EUR-Lex (europa.eu)
Depending on the root cause identified and extent of changes to be made, grouping of variations or use
of work-sharing procedures might be applicable: https://www.ema.europa.eu/en/human-
regulatory/post-authorisation/variations/worksharing-questions-answers.
The potential presence of nitrosamines must be evaluated as part of the MAA as follows:
− For the risk evaluation, Applicants are required to follow the principles for step 1 as per Q&A 2.
The risk evaluation should be submitted as an attachment to Module 1 with a corresponding
reference in Module 3.2 of the marketing authorisation dossier. To supplement the detailed risk
evaluation, the template located on the CMDh nitrosamine website (section “For additional
specific information related to nationally authorised products (including MRP/DCP)”) could also
be submitted: https://www.hma.eu/human-medicines/cmdh/nitrosamine-impurities.html. The
template is optional for CAPs. For NAPs, and DCPs, the template is mandatory and the CMDh
practical guidance located in the same section of the same website should be followed.
− If a risk of presence of nitrosamines in the medicinal product is identified, applicants are required
to provide the risk assessment outlining the impact on the benefit-risk balance of the product
− In case applicants have not submitted a risk evaluation and, if applicable, confirmatory testing
plans with their MAA, these should be submitted during the marketing authorisation review
procedure.
− If the risk evaluation was not submitted as part of the MAA, it will be requested during the MA
review process. Risk evaluation will have to be adequately documented and, if applicable,
supported by confirmatory testing in case a possible risk of presence of nitrosamines has been
identified. This information should be submitted as part of the responses to the list of questions.
− If the applicant is not able to provide satisfactory information and justification of a favourable
benefit-risk profile of the product at this stage, a request to further assess the risk of presence
of nitrosamine will be part of the further list of questions / outstanding issues depending on the
stage of the MA procedure.
− Any outstanding issues related to the quality requirements of the product would have to be
addressed before the final opinion on the granting of the MA.
For new and on-going marketing authorisation applications, the number of batches to be tested as part
of any confirmatory testing should be commensurate with the risk in line with ICH M7(R2) guideline. The
source of risk has to be well understood (e.g. by spike and purge studies) such that impurity levels are
expected to be consistent from batch to batch. Test results from a minimum of 6 pilot scale batches or
3 production scale batches may be sufficient. Depending on the risk factors for nitrosamine presence,
e.g. with risk factors being closer to the FP, more batches may need to be tested. If multiple
manufacturers, manufacturing processes and/or sources of at-risk raw materials are used, (or were used
historically during development), then testing of additional batches would be necessary to cover these
risk factors.
If a product is available in multiple strengths of the same dosage form with the same risk factors
applicable to each, then testing could be rationalised by testing only the worst-case scenario strength.
The worst-case approach should be justified by the MAH on a case-by-case basis.
During development of an analytical method, a reference standard of the relevant nitrosamine impurity
is generally needed. If, despite extensive efforts, it becomes apparent that the relevant nitrosamine
impurity cannot be synthesised, then this could be an indication that the nitrosamine either does not
exist or that there is no risk of it being formed. In such cases, it may not be necessary to conduct
confirmatory testing. This should be justified thoroughly on a case-by-case basis according to appropriate
scientific principles. The justification could include relevant literature, information on structural/stereo-
electronic features and reactivity of the parent amine, stability of the nitrosamine and experimental data
to illustrate the efforts made to synthesise and to analyse the impurity. The justification should be
included in the submitted risk assessment.
For products where nitrosamine impurities can be controlled according to ICH Q3A/B principles, see Q&A
10, confirmatory testing is generally not needed if the risk can be sufficiently mitigated based on scientific
considerations that demonstrate that the relevant ICH Q3A/B thresholds will not be exceeded. In such
cases, the justification should be documented in the risk assessment in the MAH's pharmaceutical quality
system. For products with only advanced cancer indications as defined within the scope of ICH S9, and
where the above principle has been applied to mitigate the potential risk of nitrosamine impurities, the
approach should be revised should the indication ever move beyond the relevant ICH S9 setting. In these
The control point (finished product, API or an intermediate) for nitrosamines should be selected in such
a way that it will give assurance of presence of the impurity below the acceptable limit based on
acceptable intake (AI) in the finished product. Testing is usually expected to be carried out in the finished
product, however if the source of a nitrosamine impurity is identified in the active substance
manufacturing process, control options 1 to 3 as stated in ICH M7(R2) guideline could be used to
demonstrate that the nitrosamine will not be present above the acceptable limit based on AI in the
finished product. Testing of raw materials (e.g. excipients) should also be considered if these are
potential sources of nitrosamine impurities. Exceptions from routine testing may be possible, if the root
cause of contamination is demonstrated to be well-understood:
• Only if the amount of nitrosamine present is consistently below 10% of the acceptable limit based
on AI in the API or in the finished product, then a test for the nitrosamine could be omitted from the
specification.
• Only if levels of a single nitrosamine are consistently below 30% of the acceptable limit based on AI
in the API or the finished product, skip-testing according to the ICH Q6A definition could be
acceptable.
Where nitrosamine impurities can be controlled according to ICH Q3A/B principles and the risk can be
sufficiently mitigated based on scientific considerations that demonstrate that the relevant ICH Q3A/B
thresholds will not be exceeded, then a specification limit in the dossier is not expected.
16. What are the responsibilities of MAHs for APIs with CEPs
or ASMFs?
MAHs/Applicants, manufacturing authorisation holders and API manufacturers should work together and
take precautionary measures to mitigate the risk of presence of nitrosamines during the manufacture
and storage of all medicinal products containing chemically synthesised APIs.
MAHs/Applicants must ensure that appropriate and robust risk evaluations are carried out by the relevant
manufacturing authorisation holders and API manufacturers (including ASMF or CEP holders) in
accordance with Article 46 of Directive 2001/83/EC.
APIs documented in CEPs and ASMFs may be used in different finished products, potentially with different
formulations, manufacturing processes and indications. To mitigate levels of nitrosamines impurities , a
given finished product may need a particular quality of API with additional tests or tighter limits compared
to the Ph. Eur. monograph of that substance, e.g. tighter limits for amine impurities that may be
converted to nitrosamines during formulation and storage of the finished product. It is the responsibility
of the MAH to work with its API suppliers to ensure that API of acceptable quality is used for a given
The recommendations set forward include new or additional guidance on areas such as the control of
impurities (including cohort of concern compounds), Good Manufacturing Practice, the roles and
responsibilities of manufacturers and MAHs/Applicants but also proposals for improvement of
communication with patients and healthcare professionals and cooperation with international partners.
The full recommendations are available on EMA’s website. The European medicines regulatory network
will develop an implementation plan and then work with the parties that will implement each action.
It should be noted that the lessons learnt exercise outcome has been taken into account in the Article
5(3) procedure. The implementation of recommendations of the lessons learnt exercise will strengthen
the regulatory framework and complement the outcome of this Article 5(3) procedure which provides
the scientific opinion on the presence of nitrosamine impurities in human medicines.
Nevertheless, in some exceptional cases questions on the presence of nitrosamines in the product may
be raised if a potential risk is identified during the assessment.
Chapter 3.2 provides a description on how regulators will approach the outcome from the call for review
in accordance with the different scenarios reported by MAHs.
In case of identification of one or more N-nitrosamine exceeding the AI in the finished product, or in case
that the sum of all detected N-nitrosamines exceeds the 1 in a 100,000 lifetime risk (scenario A), the
following steps are taken in order to protect public health and ensure availability of critical medicines:
• A lead authority is identified as responsible for reviewing the information available and for providing
the (preliminary) assessment of the case. The lead authority is selected as outlined in chapter 5.1.
• The Rapid Alert Network (RAN) and the availability Single Point Of Contacts (SPOCs) are informed in
order to determine the criticality of the product (in accordance with Criteria for classification of critical
medicinal products for human and veterinary use).
• The feedback from RAN and availability SPOCs is taken into account by the lead authority when
providing the preliminary recommendations on any interim or eventual required market actions and
on the acceptability of corrective and preventive actions proposed by the MAH.
• The Incident Review Network (IRN) is consulted in order to facilitate the exchange of information
and to evaluate whether additional measures are needed or whether a different regulatory pathway
is warranted.
• If market actions are recommended, each National Competent Authority (NCA) will follow up in
accordance with their national procedures and depending on the criticality of the product for their
markets.
• The use of an interim limit based on the LTL approach during CAPA implementation, as described in
Q&A 22, may be considered, as applicable, by the lead authority and NCAs on a temporary basis for
market action purposes. Please refer to chapter 3.2.1.1 of the regulatory process dealing with the
outcomes of the call for review referenced above.
• CAPA implementation timeline of up to 3 years from the establishment and publication of the AI
(nevertheless MAHs are expected to expedite CAPAs implementation).
*In any case the limit should not exceed 1.5 µg/day unless the established AI (Table 1, Q&A10) is > 1.5 µg/day or
the nitrosamine concerns a category 5 according to CPCA or the nitrosamine is shown to be negative in an enhanced
Ames test (EAT).
The approach is not applicable to the below instances where other approaches may be considered on a
case-by-case basis in consultation with the appropriate regulatory authority:
• CAPA implementation exceeding 3 years from the establishment and publication of the AI;
• New/ongoing regulatory applications.
The above interim limits are based on the LTL approach outlined in the ICH M7 guideline, using the two
most conservative adjustment factors (6.7 and 13.3 x AI). The application of these adjustment factors
would not be expected to exceed a theoretical excess cancer risk of 1 in 100,000 during the period of
CAPA implementation.
MAHs are expected to ensure that the implementation of adequate controls for the detected nitrosamines
is done as a matter of priority. During the use of the interim limit, monitoring measures may be evaluated
by the lead authority as required. However, it is not the expectation that MAHs include these interim
limits in specifications via variation.