Skip to content
biopharma.ai
  • Home
  • Design
  • Produce
  • Test
  • News
  • Docs
  • Events
    • Webinars
  • About
Contact Us
Contact Us
biopharma.ai
  • Home
  • Design
  • Produce
  • Test
  • News
  • Docs
  • Events
    • Webinars
  • About

Docs

  • ICH guidlines
    • Safety Guidelines
      • S10
      • S11
      • S12
      • S9
      • S8
      • S7B
      • S7A
      • S6(R1)
      • S5(R3)
      • S4
      • S3B
      • S3A
      • S2(R1)
      • S1C(R2)
      • S1B(R1)
      • S1A
View Categories
  • Home
  • Docs
  • Docs
  • ICH guidlines
  • Safety Guidelines
  • S9

S9

34 min read

NONCLINICAL EVALUATION FOR ANTICANCER PHARMACEUTICALS

1.       INTRODUCTION

1.1     Objectives of the Guideline

The purpose of this guidance is to provide information to assist in the design of an appropriate   program   of  nonclinical   studies   for   the   development   of  anticancer pharmaceuticals. The guidance provides recommendations for nonclinical evaluations to  support  the  development  of anticancer  pharmaceuticals  in  clinical trials  for the treatment of patients with advanced disease and limited therapeutic options.

This  guideline  aims  to  facilitate  and   accelerate  the   development  of  anticancer pharmaceuticals  and  to  protect  patients  from  unnecessary  adverse  effects,  while avoiding   unnecessary   use   of   animals,   in   accordance   with   the   3R   principles (reduce/refine/replace), and other resources.

As appropriate, the principles described in other ICH guidelines should be considered in   the   development   of   anticancer   pharmaceuticals.   Specific   situations   where recommendations for nonclinical testing deviate from other guidance are described in this document.

1.2     Background

Because malignant tumors are life-threatening, the death rate from these diseases is high, and existing therapies have limited effectiveness, it is desirable to provide new, effective anticancer drugs to patients more expeditiously.

There have been no internationally accepted objectives or recommendations on the design and conduct of nonclinical studies to support the development of anticancer pharmaceuticals in clinical trials for the treatment of patients with advanced disease and limited therapeutic options. Nonclinical evaluations are conducted to:

1) identify the pharmacologic properties of a pharmaceutical;

2) establish a safe initial dose level for the first human exposure; and

3) understand the toxicological profile of a pharmaceutical (e.g., identification of target organs, exposure-response relationships, and reversibility).

In the development of anticancer drugs, clinical studies often involve cancer patients whose disease condition is progressive and fatal. In addition, the dose levels in these clinical  studies  often  are  close  to  or  at  the  adverse  effect  dose  levels.  For  these reasons, the type, timing and flexibility called for in the design of nonclinical studies of anticancer pharmaceuticals can differ from those elements in nonclinical studies for other pharmaceuticals.

1.3     Scope

This guideline provides information for pharmaceuticals that  are intended to treat cancer in patients with serious and life threatening malignancies. For the purpose of this guideline, this patient population is referred to as patients with advanced cancer.

The    guideline    applies    to    both    small    molecule     and    biotechnology-derived pharmaceuticals (biopharmaceuticals), regardless of the route of administration. This guideline  describes  the  type  and  timing  of  nonclinical  studies  in  relation  to  the development   of  anticancer  pharmaceuticals  in  patients  with  advanced  cancer  and references other guidance as appropriate. It describes the minimal considerations for initial clinical trials in patients with advanced cancer whose disease is refractory or resistant to available therapy, or where current therapy is not considered to be providing benefit. The nonclinical  data  to  support  Phase  I  and  the  clinical  Phase  I  data  would  normally  be sufficient for moving to Phase II and into  second or first line therapy in patients with advanced cancer. The  guideline  also describes further non-clinical data to be collected during  continued  clinical  development  in  patients  with  advanced  cancer.  When  an anticancer pharmaceutical is further investigated in cancer patient populations with long expected survival (e.g., those administered pharmaceuticals on a chronic basis to reduce the  risk  of  recurrence  of  cancer),  the  recommendations  for  and  timing  of  additional nonclinical studies depend upon the available nonclinical and clinical data and the nature of the toxicities observed.

This  guideline  does  not  apply  to  pharmaceuticals  intended  for  cancer  prevention, treatment  of  symptoms  or  side  effects  of  chemotherapeutics,  studies  in  healthy volunteers, vaccines, or cellular or gene therapy. If healthy volunteers are included in clinical trials, the ICH M3 guideline should be followed. Radiopharmaceuticals are not covered in this guideline, but some of the principles could be adapted.

1.4     General Principles

The   development   of   each   new   pharmaceutical   calls   for   studies   designed   to characterize its pharmacological and toxicological properties according to its intended use in humans. Modification of “standard” nonclinical testing protocols generally is warranted to address novel characteristics associated with the pharmaceutical or with the manner in which it is to be used in humans.

The manufacturing process can change during the course of development. However, the  active  pharmaceutical  substance  used  in  nonclinical  studies  should  be  well characterized and should adequately represent the active substance to be used in the clinical trials.

In general, nonclinical safety studies that are used to support the development of a pharmaceutical should be conducted in accordance with Good Laboratory Practices.

2.       STUDIES TO SUPPORT NONCLINICAL EVALUATION

2.1     Pharmacology

Prior to Phase I studies, preliminary characterization of the mechanism(s) of action and  schedule  dependencies  as  well  as  anti-tumor  activity  of  the  pharmaceutical should have been made. Appropriate models should be selected based on the target and mechanism of action, but the pharmaceutical need not be studied using the same tumor types intended for clinical evaluation.

These studies can:

â—Ź    provide nonclinical proof of principle;

â—Ź    guide schedules and dose-escalation schemes;

â—Ź    provide information for selection of test species;

â—Ź   aid in start dose selection and selection of investigational biomarkers, where appropriate; and

â—Ź    if relevant, justify pharmaceutical combinations.

Understanding the secondary pharmacodynamic properties of a pharmaceutical could contribute  to  the  assessment  of safety  for  humans,  and  those  properties  might  be investigated as appropriate.

2.2     Safety Pharmacology

An  assessment  of  the  pharmaceutical’s  effect  on  vital  organ  functions  (including cardiovascular, respiratory and central nervous systems) should be available before the  initiation  of  clinical  studies;  such  parameters  could  be  included  in  general toxicology   studies.  Detailed  clinical  observations  following  dosing  and  appropriate electrocardiographic  measurements  in  non-rodents  are  generally  considered  sufficient. Conducting  stand-alone  safety  pharmacology  studies  to  support  studies  in  patients with  advanced cancer is not called for.  In cases where specific concerns have been identified  that  could  put  patients  at  significant  additional  risks  in  clinical  trials, appropriate safety pharmacology studies described in ICH S7A and/or S7B should be considered.  In the  absence  of a  specific risk,  such  studies will  not be  called for to support clinical trials or for marketing.

2.3     Pharmacokinetics

The  evaluation  of  limited  pharmacokinetic  parameters  (e.g.,  peak  plasma/serum levels,  area  under  the  curve  (AUC),  and  half-life)  in  the  animal  species  used  for nonclinical studies can facilitate dose selection, schedule and escalation during Phase I studies. Further information on absorption, distribution, metabolism and excretion of  the  pharmaceutical  in  animals  should  normally  be  generated  in  parallel  with clinical development.

2.4     General Toxicology

The primary objective of Phase I clinical trials in patients with advanced cancer is to assess the safety of the pharmaceutical. Phase I assessments can include dosing to a maximum tolerated dose (MTD) and dose limiting toxicity (DLT). Toxicology studies to determine a no observed adverse effect level (NOAEL) or no effect level (NOEL) are not considered essential to support clinical use of an anticancer pharmaceutical. As the  toxicity  of  the  pharmaceutical  can  be  greatly  influenced  by  its  schedule  of administration,  an  approximation  of  its  clinical  schedule  should  be  evaluated  in toxicology studies. This is further discussed in Section 3.3 and 3.4.

Assessment   of  the   potential   to   recover   from   toxicity   should   be   provided   to understand  whether  serious  adverse  effects  are  reversible  or  irreversible. A study that includes a terminal non-dosing period is called for if there is severe toxicity

at  approximate  clinical  exposure  and  recovery  cannot  be  predicted  by  scientific assessment.  This  scientific  assessment  can  include  the  extent  and  severity  of the pathologic lesion and the regenerative capacity of the organ system showing the effect. If  a  study  of  recovery  is   called  for,  it   should  be   available  to   support  clinical development. The demonstration of complete recovery is not considered essential.

For small molecules, the general toxicology testing usually includes rodents and non- rodents.  In  certain  circumstances,  determined  case-by-case,  alternative  approaches can be appropriate (e.g., for genotoxic drugs targeting rapidly dividing cells, a repeat- dose toxicity study in one rodent species might be considered sufficient, provided the rodent is a relevant species). For biopharmaceuticals, see ICH S6 for the number of species to be studied.

Toxicokinetic evaluation should be conducted as appropriate.

2.5     Reproduction Toxicology

An embryofetal toxicology assessment is conducted to communicate potential risk for the  developing  embryo  or  fetus  to  patients  who  are  or  might  become  pregnant. Embryofetal toxicity studies of anticancer pharmaceuticals should be available when the marketing application is submitted, but these studies are not considered essential to support clinical trials intended for the treatment of patients with advanced cancer. These   studies   are   also  not   considered   essential  for  the  purpose  of  marketing applications for pharmaceuticals that are genotoxic and target rapidly dividing cells (e.g., crypt cells, bone marrow) in general toxicity studies or belong to a class that has been well characterized as causing developmental toxicity.

For  small  molecules,  embryofetal  toxicology  studies  are typically  conducted  in  two species  as  described by ICH  S5(R2).  In  cases where  an embryofetal developmental toxicity  study is  positive for  embryofetal lethality or teratogenicity,  a  confirmatory study in a second species is usually not warranted.

For  biopharmaceuticals,  an  assessment  in  one  pharmacologically  relevant  species should usually be sufficient. This assessment might be done by evaluating the toxicity during  the  period  of  organogenesis   or  study  designs  as  described  by   ICH  S6. Alternative approaches might be considered appropriate if scientifically justified. The alternative approaches might include a literature assessment, assessment of placental transfer, the direct or indirect effects of the biopharmaceutical, or other factors.

A  study  of fertility  and  early  embryonic  development  is  not  warranted  to  support clinical  trials  or  for  marketing  of pharmaceuticals  intended  for  the  treatment  of patients with advanced cancer. Information available from general toxicology studies on the pharmaceutical’s effect on reproductive organs should be used as the basis of the assessment of impairment of fertility.

A pre- and postnatal toxicology study is generally not warranted to support clinical trials or for marketing of pharmaceuticals for the treatment of patients with advanced cancer.

2.6     Genotoxicity

Genotoxicity   studies   are   not   considered   essential  to   support   clinical  trials  for therapeutics  intended to treat  patients with  advanced  cancer.  Genotoxicity  studies

should be performed to support marketing (see ICH S2). The principles outlined in ICH S6 should be followed for biopharmaceuticals. If the in vitro assays are positive, an in vivo assay might not be warranted.

2.7     Carcinogenicity

The appropriateness of a carcinogenicity assessment for anticancer pharmaceuticals is described   in   ICH   S1A.   Carcinogenicity   studies   are   not   warranted   to   support marketing for therapeutics intended to treat patients with advanced cancer.

2.8     Immunotoxicity

For most anticancer pharmaceuticals, the design components of the general toxicology studies  are  considered  sufficient  to  evaluate  immunotoxic  potential  and  support marketing.  For  immunomodulatory  pharmaceuticals,  additional  endpoints  (such  as immunophenotyping by flow cytometry) might be included in the study design.

2.9     Photosafety testing

An initial assessment of phototoxic potential should be conducted prior to Phase I, based   on photochemical properties of the drug and information on other members in the class. If assessment of these data indicates a potential risk, appropriate protective measures should betaken during outpatient trials. If the photosafety risk cannot be adequately evaluated

based on nonclinical data or clinical experience, a photosafety assessment consistent with the principles described in ICH M3 should be provided prior to marketing.

3.        NONCLINICAL DATA TO SUPPORT CLINICAL TRIAL DESIGN AND MARKETING

3.1     Start Dose for First Administration in Humans

The  goal  of selecting  the  start  dose  is  to  identify  a  dose  that  is  expected  to  have pharmacologic  effects  and  is  reasonably  safe  to  use.  The   start  dose  should  be scientifically justified  using  all  available  nonclinical  data  (e.g.,  pharmacokinetics, pharmacodynamics, toxicity), and its selection based on various approaches (see Note 2).  For most  systemically  administered  small molecules, interspecies  scaling of the animal doses to an equivalent human dose is usually based on normalization to body surface area. For both small molecules and biopharmaceuticals, interspecies scaling based on body weight, AUC, or other exposure parameters might be appropriate.

For biopharmaceuticals with immune agonistic properties, selection of the start dose using a minimally anticipated biologic effect level (MABEL) should be considered.

3.2     Dose Escalation and the Highest Dose in a Clinical Trial

In general, the highest  dose or exposure tested in the nonclinical  studies  does  not limit the  dose-escalation  or highest  dose investigated in  a  clinical trial in patients with  cancer.  When  a  steep  dose-  or  exposure-response  curve  for  severe  toxicity  is observed  in  nonclinical  toxicology  studies,  or  when  no  preceding  marker  of severe

toxicity is available, smaller than usual dose increments (fractional increments rather than dose doubling) should be considered.

3.3    Duration   and   Schedule   of  Toxicology   Studies   to   Support   Initial Clinical Trials

In Phase I clinical trials, treatment can continue according to the patient’s response, and  in  this  case,  a  new  toxicology  study  is  not  called  for  to  support  continued treatment beyond the duration of the completed toxicology studies.

The  design  of nonclinical  studies  should  be  appropriately  chosen  to  accommodate different  dosing  schedules  that  might  be  utilized  in  initial  clinical trials.  It  is  not expected that the exact clinical schedule always will be followed in the toxicological study, but the information provided from the toxicity studies should be sufficient to support the clinical dose and schedule and to identify potential toxicity. For example, one factor that can be considered is the half-life in the test species and the projected (or  known)  half-life  in  humans.  Other  factors  could  include  exposure  assessment, toxicity profile, saturation of receptors, etc. Table 1 provides examples of nonclinical treatment   schedules   that    are   commonly   used   in    anticancer   pharmaceutical development  and  can be  used  for  small  molecules  or  biopharmaceuticals.  In  cases where  the  available  toxicology  information  does  not  support  a  change  in  clinical schedules, an additional toxicology study in a single species is usually sufficient.

3.4    Duration    of   Toxicology   Studies    to   Support    Continued   Clinical

Development and Marketing

The nonclinical data to support Phase I and the clinical Phase I data would normally be  sufficient for moving to Phase II and into  second or first line therapy in patients with advanced cancer. In support of continued development of an anticancer pharmaceutical  for  patients  with  advanced  cancer,  results  from  repeat  dose  studies  of 3  months’ duration following the intended clinical schedule should be provided prior to initiating Phase III studies. For most pharmaceuticals intended for the treatment of patients  with  advanced  cancer,  nonclinical  studies  of  3  months  duration  are  considered  sufficient to support marketing.

When  considering  a  change  in  the  clinical  schedule,  an  evaluation  of the  existing clinical data should be conducted to justify such change. If the clinical data alone are inadequate to  support the  change in  schedule, the factors  discussed in  Section  3.3 above should be considered.

3.5     Combination of Pharmaceuticals

Pharmaceuticals planned for use in combination should be well studied individually in toxicology  evaluations.  Data  to  support  a  rationale  for  the  combination  should  be provided   prior   to   starting   the   clinical   study.   In   general,   toxicology   studies investigating the safety of combinations of pharmaceuticals intended to treat patients with  advanced  cancer  are  not  warranted.  If  the  human  toxicity  profile  of  the pharmaceuticals   has   been   characterized,   a   nonclinical    study   evaluating   the combination  is  not  usually  warranted.  For  studies  in  which  at  least  one  of these compounds is in early stage development (i.e., the human toxicity profile has not been characterized),  a  pharmacology  study  to  support  the  rationale for the  combination should be provided. This study should provide evidence of increased activity in the

absence of a substantial increase in toxicity on the basis of limited safety endpoints, such as mortality, clinical signs, and body weight. Based on available information, a determination  should  be  made  whether  or  not  a  dedicated  toxicology  study  of the combination is warranted.

3.6    Nonclinical Studies to Support Trials in Pediatric Populations

The general paradigm for investigating most anticancer pharmaceuticals in pediatric patients is first to define a relatively safe dose in adult populations and then to assess some fraction of that dose in initial pediatric clinical studies. The recommendations for  nonclinical  testing  outlined  elsewhere  in  this  document  also  apply  for  this population. Studies in juvenile animals are not usually conducted in order to support inclusion of pediatric populations for the treatment of cancer. Conduct of studies in juvenile  animals  should be  considered  only when human  safety  data  and  previous animal  studies  are  considered  insufficient  for  a  safety  evaluation  in  the  intended pediatric age group.

4.       OTHER CONSIDERATIONS

4.1     Conjugated Products

Conjugated products are pharmaceuticals covalently bound to carrier molecules, such as proteins, lipids, or  sugars. The safety of the conjugated material is the primary concern. The safety of the unconjugated material, including the linker used, can have a more limited evaluation. Stability of the conjugate in the test species and human plasma   should  be  provided.  A  toxicokinetic  evaluation   should   assess  both  the conjugated  and  the  unconjugated  compound  after  administration  of the  conjugated material.

4.2     Liposomal Products

A complete evaluation of the liposomal product is not warranted if the unencapsulated material has been well characterized. As appropriate, the safety assessment should include a toxicological evaluation of the liposomal product and a limited evaluation of the  unencapsulated  pharmaceutical  and  carrier  (e.g.,  a  single  arm  in  a  toxicology study).  The  principle  described  here  might  also  apply  to  other  similar  carriers.  A toxicokinetic evaluation should be conducted as appropriate. If possible, such an evaluation should assess both the liposomal product and the free compound after administration of the liposomal product.

4.3     Evaluation of Drug Metabolites

In  some  cases,  metabolites  that  have  been  identified  in  humans  have  not  been qualified  in  nonclinical  studies.  For  these  metabolites,  a  separate  evaluation  is generally not warranted for patients with advanced cancer.

4.4     Evaluation of Impurities

It  is  recognized  that  impurity  standards  have  been  based  on  a  negligible  risk,  as discussed  in  ICH  Q3A  and  Q3B.  Exceeding  the  established  limits  for  impurities identified    in    these    ICH    guidelines    could    be    appropriate    for    anticancer

pharmaceuticals, and a justification should be provided in the marketing application. The justification could include the disease being treated and the patient population, the nature of the parent pharmaceutical (pharmacologic properties, genotoxicity and carcinogenic  potential,  etc.),  duration  of  treatment,  and  the  impact  of  impurity reduction  on  manufacturing.  Further,  the  qualification  assessment  could  include consideration of either the dose or concentration tested in nonclinical study relative to clinical  levels.  For  genotoxic  impurities,  several  approaches  have been  used  to  set limits based on increase in lifetime risk of cancer. Such limits are not appropriate for pharmaceuticals intended to treat patients with advanced cancer, and justifications described  above  should be considered to set higher limits.  Impurities that  are  also metabolites   present   in   animal   and/or   human   studies   are   generally   considered qualified.

Table  1: Examples of Treatment Schedules for Anticancer Pharmaceuticals to Support Initial Clinical Trials

Clinical ScheduleExamples of Nonclinical Treatment Schedule1,2,3,4
Once every 3-4 weeksSingle dose
Daily for 5 days every 3 weeksDaily for 5 days
Daily for 5-7 days, alternating weeksDaily for 5-7 days, alternating weeks (2-dose cycles)
Once   a   week   for   3   weeks,1 week offOnce a week for 3 weeks
Two or three times a weekTwo or three times a week for 4 weeks
DailyDaily for 4 weeks
WeeklyOnce a week for 4-5 doses

1  Table 1 describes the dosing Phase. The timing of the toxicity assessment(s) in the non-clinical studies should be scientifically justified based on the anticipated toxicity profile and the clinical schedule. For example, both a sacrifice shortly after the dosing Phase to examine early toxicity and a later sacrifice to examine late onset of toxicity should be considered.

2    For  further  discussion  regarding  flexibility  in  the  relationship  of  the  clinical schedule and the non-clinical toxicity studies, see Section 3.3.

3  The treatment schedules described in the table do not specify recovery periods (see Section 2.4 and Note 1 regarding recovery).

4  The treatment schedules described in this table should be modified as appropriate for molecules with extended pharmacodynamic effects, long half-lives or potential for anaphylactic reactions. In addition, the potential effects of immunogenicity should be considered (see ICH S6).

5.       NOTES

1. For nonrodent studies, dose groups usually consist of at least 3 animals/sex/group, with an additional 2/sex/group for recovery, if appropriate (see Section 2.4). Both sexes should generally be used, or justification should be given for specific omissions.

2. A common approach for many small molecules is to set a start dose at  1/10 the Severely Toxic Dose in 10% of the animals (STD 10) in rodents. If the non-rodent is the most appropriate species, then 1/6 the Highest Non-Severely Toxic Dose (HNSTD) is considered an appropriate starting dose. The HNSTD is defined as the highest dose level  that   does   not   produce   evidence   of  lethality,   life-threatening  toxicities   or irreversible findings.

What are your Feelings
Share This Article :
  • Facebook
  • X
  • LinkedIn
  • Pinterest
Still stuck? How can we help?

How can we help?

Updated on March 3, 2025
S12S8

Powered by BetterDocs

Leave a Reply Cancel reply

Your email address will not be published. Required fields are marked *

Connect

Copyright © 2025 Aibio AB

biopharma.ai
Manage Consent
To provide the best experiences, we use technologies like cookies to store and/or access device information. Consenting to these technologies will allow us to process data such as browsing behavior or unique IDs on this site. Not consenting or withdrawing consent, may adversely affect certain features and functions.
Functional Always active
The technical storage or access is strictly necessary for the legitimate purpose of enabling the use of a specific service explicitly requested by the subscriber or user, or for the sole purpose of carrying out the transmission of a communication over an electronic communications network.
Preferences
The technical storage or access is necessary for the legitimate purpose of storing preferences that are not requested by the subscriber or user.
Statistics
The technical storage or access that is used exclusively for statistical purposes. The technical storage or access that is used exclusively for anonymous statistical purposes. Without a subpoena, voluntary compliance on the part of your Internet Service Provider, or additional records from a third party, information stored or retrieved for this purpose alone cannot usually be used to identify you.
Marketing
The technical storage or access is required to create user profiles to send advertising, or to track the user on a website or across several websites for similar marketing purposes.
Manage options Manage services Manage {vendor_count} vendors Read more about these purposes
View preferences
{title} {title} {title}