Nanopharmaceuticals-iEvaluation Guidelines Draft

Download as pdf or txt
Download as pdf or txt
You are on page 1of 49

GUIDELINES

FOR EVALUATION OF
NANOPHARMACEUTICALS
IN INDIA

Jointly Prepared by:


Department of Biotechnology, GoI
Indian Society of Nanomedicine
Guidelines for Evaluation of
Nanopharmaceuticals
in India

Jointly prepared by

Department of Biotechnology, GoI


Indian Society of Nanomedicine (ISNM)

EDITORS

Y. K. Gupta G. N. Singh A.K. Dinda A.K. Pradhan

AIIMS CDSCO AIIMS CDSCO


Guidelines on Nanopharmaceuticals

Experts Institute/ Organization


Dr. Y. K. Gupta: All India Institute of Medical Sciences, New Delhi

Dr. Amit K. Dinda: All India Institute of Medical Sciences, New Delhi

Dr. Gagandeep Kang: Translational Health Science and Technology Institute

Mr. A. K. Pradhan: Central Drug Standard Control Organization


Dr. C. Nath: Central Drug Research Institute, Lucknow

Dr. Manzoor: Amrita Institute of Medical Sciences, Kochi


Postgraduate Institute of Medical Education and Research,
Dr. Bikash Medhi:
Chandigarh
Dr. Rajkumar: Adyar Cancer Institute
Dr. Ramteke: Clinical Development Services Agency

Dr. Sucheta Kurundkar: Clinical Development Services Agency

Dr. Dhananjay Tiwari: Department of Biotechnology


Dr. Suchita Ninawe: Department of Biotechnology
Jawaharlal Nehru Centre for Advanced Scientific Research,
Dr. Tapas Kundu:
Bangalore
Dr. Arun Chattopadhya: Indian Institute of Technology, Guwahati

Dr. Deepthi Menon: Amrita Institute of Medical Sciences, Kochi

Dr. Geeta Jotwani: Indian Council of Medical Research


Dr. Namrata Pathak: Department of Science and Technology

AIIMS Team

Madhusudan Bhat: Coordinator

2
Guidelines on Nanopharmaceuticals

Contents

S No. Title Page no.


1 Abbreviations

2 Introduction

3 Scope of the Guidelines

4 General Considerations of the Guidelines

5 Nanopharmaceuticals: Definition and Categorization

6 Scientific Rationality of Making the Nanopharmaceuticals


Specific considerations for Evaluation of Nanopharmaceuticals in the
7
context of Schedule Y of Drugs and Cosmetics Rules 1945

8 Stability Testing of Nanopharmaceuticals

9 Animal Pharmacology Data

10 Animal Toxicology Data

11 Clinical Trials Data

12 Information Required for Evaluation of Nanopharmaceuticals


13 Pharmacovigilance of Nanopharmaceuticals
14 Conclusion

15 Glossary

3
Guidelines on Nanopharmaceuticals

1. Abbreviations

ADME: Absorption, Distribution, Metabolism, Excretion

API: Active Pharmaceutical Ingredient

AUC: Area Under Curve

CDSCO: Central Drug Standard Control Organization

D & C Act: Drugs and Cosmetics Act, 1940

D & C Rules: Drugs and Cosmetics Rules, 1945

DCG(I): Drugs Controller General (India)

DLS: Dynamic Light Scattering

DLT: Dose Limiting Toxicities

EU: European Union

FDA: United States Food and Drug Administration

GLP: Good Laboratory Practice

HED: Human Equivalent Dose

ICH: International Conference on Harmonization of Technical


Requirements for Registration of Pharmaceuticals for Human Use

IND: Investigational New Drug

MTD: Maximum Tolerated Dose

NCE: New Chemical Entity

OECD: Organization for Economic Co-operation and Development

PEG: Polyethylene glycol

4
Guidelines on Nanopharmaceuticals

PIM: Pulmonary Intravascular Macrophage

PK/PD: Pharmacokinetics/ Pharmacodynamics

PLA: Polylactic acid

PLGA: Poly lactic-co-glycolic acid

RES: Reticulo-Endothelial System

SoPs: Standard Operating procedures

5
Guidelines on Nanopharmaceuticals

2. Introduction

Nanoscience is the study of materials which are in nanoscale range.

Conversion of any material in nanoscale results in alteration of its

physicochemical, biological, mechanical, optical, electronic, etc. properties. These

newly acquired (novel) properties of the materials due to conversion into a

nanoscale can be utilized for different useful activities. Thus, it is an enabling

technology, relevant for diverse sectors, such as chemicals, consumer products,

health, energy, various other industries and the environment. The use of this

technology is increasing exponentially in the pharmaceutical sector.

Nanopharmaceutical is an emerging field that combines nanotechnology with

pharmaceutical and biomedical science with the goal of targeted drug delivery

which may improve efficacy and safety profile. The concept of 5Rs: ‘right

target/efficacy’, ‘right tissue/exposure’, ‘right patients’, ‘right safety’, and ‘right

commercial potential’ as postulated by Cook D et al (2014) may help in successful

development of nanopharmaceuticals.

Alteration of the substance into nanoscale associated with drug delivery may also

significantly alter the pharmacokinetic, biodistribution and toxicokinetic

parameters of the conventional/traditional drugs raising various concerns related to

quality, safety and efficacy of the nanopharmaceutical products.

6
Guidelines on Nanopharmaceuticals

Efforts have been made for developing regulatory guidelines for

nanopharmaceuticals in different countries. Since, there are no specific guidelines

for development and evaluation of nanopharmaceuticals in India, it has been felt

necessary to formulate comprehensive guidelines focusing on the quality, safety

and efficacy of nanopharmaceutical for therapeutic use. These guidelines are

intended to provide transparent, consistent and predictable regulatory pathways for

nanopharmaceuticals in India.

Nanotechnology is an enabling technology for various incremental and disruptive

innovations. Application of this technology has tremendous potential in

pharmaceutical industry where it can improve the therapeutic efficacy with

targeted delivery of the drug to the site of disease. There may be a concurrent

reduction of the dose of the drug with lowering of toxicity.

However, the nanocarriers/ nanopharmaceuticals have a higher tendency of tissue

sequestration which alters the PK/PD of the conventional/traditional drug which is

loaded in the nanosystems. This may lead to additional risk of tissue based toxicity

with low serum concentration.

Considering the complexity of the nanomaterial behavior in the biological

environment certain degree of uncertainty may be inherent to such system.

7
Guidelines on Nanopharmaceuticals

These guidelines have the aim to ensure the quality, safety and efficacy as well as

encourage the commercialization of nanotechnology based innovation with high

benefit and low risk ratio.

There are no uniform internationally acceptable guidelines for

nanopharmaceuticals.. The usual concensus for evaluation of quality, safety and

efficacy of nanotechnology based products is to have a ‘case by case approach’

taking into consideration of the physical, chemical and biological characteristics of

the nanoparticle used and the product, route of administration, the indication for

which the product is intended to be used and other related aspects.

8
Guidelines on Nanopharmaceuticals

3. Scope of the Guidelines

These guidelines apply to the nanopharmaceuticals in the form of finished

formulation as well as API of a new molecule or an already approved molecule

with altered dimensions, properties or phenomenon associated with the application

of nanotechnology intended to be used for diagnosis, treatment, mitigation or

prevention of diseases in human.

These guidelines do not apply to the conventional drug with incidental presence of

nanoparticles or drug products containing microorganisms or proteins which are

naturally present in the nanoscale range.

These are also not applicable to medical devices, in-vitro diagnostics, tissue

engineered product using nanotechnology and nano particle modified cell based

therapy.

9
Guidelines on Nanopharmaceuticals

4. General Considerations of the Guidelines

Safety studies should be conducted as per general guidelines specified in Schedule

Y of Drugs and Cosmetics Rules, 1945. However, in case any specific study is not

included in the Schedule Y, the principles of ICH guidelines for pharmaceuticals or

OECD guidelines for chemicals may be followed. This document may also serve

as useful guidelines for manufacturers, importers of nanopharmaceuticals and other

stakeholders involved in research and development of nanopharmaceuticals.

These guidelines are in conformity with the provisions of Drugs and Cosmetics

Act, 1940 and Rules,1945as amended from time to time, with certainspecific

aspects of quality, safety and efficacy applicable to nanopharmaceuticals.

These guidelines have evolved with consideration of the following documents

Schedule Y of D & C Rules, 1945

Guidance for Industry Considering Whether an FDA-Regulated Product Involves

the Application of Nanotechnology, 2014

Second Regulatory Review on Nanopharmaceuticals, European Union, 2012

Regulatory Aspects of the Nanopharmaceutical in the EU, 2017

In these guidelines, the nanopharmaceuticals have been classified according to

their degradability, organicity, function and status of approval. Accordingly, the

safety and efficacy data requirements have been described.

10
Guidelines on Nanopharmaceuticals

Specific scientific evidence required for approval of any nanopharmaceutical and

the strategies for pharmacovigilance of such products have also been incorporated

in these guidelines.

Each application should be considered on its own merit of the data submitted using

scientific judgement and logical argument.

For new generation of nanomaterials, development of methods for safety testing

and risk assessment, and a better availability of quality data on nanomaterials for

regulatory purposes are essential.

11
Guidelines on Nanopharmaceuticals

5 Nanopharmaceuticals: Definition and Categorization

5.1 Definition

A nanopharmaceutical is defined as a pharmaceutical preparation containing

nanomaterials intended for internal or external application on the body for the

purpose of therapeutics, diagnostics and any health benefit.

These are the products that contain materials in the size scale range of 1 to 100nm

in at least in one dimension. However, if the particle size is >100nm and <1000

nm, it will also fall within the definition, provided it has altered or different

pharmaceutical characteristics associated with application of nanotechnology

compared with API1,2.

The size distribution of the nanopharmaceutical: Not less than 1% beyond the nano

particle range 1 to 1000 nm is permitted. Further, the particles should be in the

claimed nano size range. At any time point during the claimed stability period, the

particle size range should not decline/ alter >10 %.

12
Guidelines on Nanopharmaceuticals

5.2 Categorization

Nanopharmaceuticals may be categorized depending on the nature and functions of

the nanomaterial as well as the approval status of the nanomaterial and the

conventional non nano form of the drug. Accordingly, nanopharmaceuticals are

categorized as under-

I According to degradability of nanomaterial

The basic difference between biodegradable and non-biodegradable is that

biodegradable items decompose or break down naturally. Non-

biodegradable items do not.

1. Biodegradable

Biodegradable nanoparticles have been used frequently as drug delivery

vehicles due to its improved bioavailability, better encapsulation, control

release and reduction of toxic potential. Examples of biodegradable

nanoparticles are PEG, albumin, PLA, PLGA, chitosan, gelatin,

polycaprolactone, poly-alkyl-cyanoacrylates, etc.

2. Nonbiodegradable

Nonbiodegradable nanoparticles are relatively less used in pharmaceutical

products (though these systems are more commonly used in cosmeceuticals).

Almost all non-biodegradable nanoparticles have potential to cause

13
Guidelines on Nanopharmaceuticals

cytotoxic effects of particle due to long sequestration without significant

degradation and excretion. Some examples of non biodegradable

nanoparticles are titanium oxide, iron oxide, and metals such as gold, silver,

platinum, etc.

II According to nature of nanomaterial

Nanomaterial may be organic or inorganic in nature. The composition and

fabrication methods will determine the properties of nanoparticles. It may

also be multicomponent nanoparticle.

1. Organic Nanoparticles

These are the nanomaterials or nanoparticles composed of

organic compounds like lipids, proteins, carbohydrates. They have been

primarily developed for drug delivery to reduce or overcome the risk of

toxicity due to the intracellular and/or tissue sequestration there by increased

bioavilability at the site of action.

Examples of organic nanoparticles used in pharmaceutical formulations are

liposome, albumin, polymer–protein, or polymer–drug conjugates. The

molecules used for the fabrication of the organic nanoparticles are usually

biodegradable which make them the most appealing systems for drug

14
Guidelines on Nanopharmaceuticals

delivery and biomedical applications. However, they may have limited

chemical and mechanical stability.

2. Inorganic Nanoparticles

The inorganic nanoparticles are generally composed of an inorganic

component. Depending on the composition, shape, size, surface property and

crystallinity, these nanoparticles may have a number of tunable physical

properties, such as optical absorption (e.g., metallic nanoparticles),

fluorescence (semiconductor quantum dots), and magnetism (e.g., iron

oxides).

Inorganic nanoparticles are more stable than organic nanostructures.

Inorganic nanoparticles may have several advantages over organic ones.

They are easier to prepare with a defined size and a very narrow size

distribution. More interestingly, they often exhibit multiple useful functions,

for example, as heat generation and contrast function for imaging. However,

most of the inorganic nanoparticles may not be biodegradable with a

potential for long term sequestration and toxicity.

3. Multicomponent nanoparticles

These are the nanoparticles composed of two or more different materials.

The integration of multiple materials in one structure offers opportunities for

15
Guidelines on Nanopharmaceuticals

enhanced physical and chemical properties and for targeting drug delivery

along with many other useful functions within a single nanostructure.

However, stabilization of multiple materials within the nanostructure is

challenging. For example, magnetic liposomes containing an aqueous

dispersion of iron oxide incorporated on the lipid surface.

III According to nanoform of the ingredient

1. Nanocarriers loaded with Active Pharmaceutical Ingredient (API)

A nanocarrier is a nanomaterial being used as a transport module for another

substance like a drug. Common examples include micelles, polymer

conjugates, polymeric nanoparticles, carbon-based materials (carbon

nanotubes), lipid-based carriers (liposomes, micelles), dendrimers, gold

nanoparticles (nanoshells, nanocages), etc.

Examples of drugs loaded with nanocarriers are liposomal amphotericin B,

albumin bound paclitaxel, liposomal doxorubicin, etc.

Because of their small size, nanocarriers can deliver drugs to otherwise

inaccessible sites around the body. These also have the advantage of targeted

drug delivery to specific sites. In the area of cancer nanomedicine the

nanoparticles are designed to exploit the Enhanced Permeability and

16
Guidelines on Nanopharmaceuticals

Retention (EPR) effect in the tumor tissue which is particularly helpful in

enhancing therapeutic index and lowering of off target toxicity.

2. APIs converted to nano form

Some of the conventional/traditional drugs may be converted into

nanocrystals, thereby increasing their potential for improved dissolution and

bioavailability. Examples are sirolimus, tacrolimus, fenofibrate, cyclosporin,

griseofulvin, etc.

IV According to the approval status of drug and nanomaterial

Based on the approval status of drug and the nanomaterial, the requirements

of quality, safety and efficacy data may vary. All nanopharmaceutical

preparations will be treated as Investigational New Drug (IND) permanently.

They may be subject to differential scrutiny according to the following

categories:

1. The drug is a new molecular entity and the nanocarrier is also new and not

approved in the country. Such product would be treated as Investigational

New Drug (IND) and the general requirement for quality, safety and efficacy

will be required to be undertaken as specified in Schedule Y of Drugs &

17
Guidelines on Nanopharmaceuticals

Cosmetics Rules, 1945.

18
Guidelines on Nanopharmaceuticals

2. The drug is a New Molecular Entity but the nanocarrier system is already

used for other nanopharmaceutical. Such product should be considered as an

Investigational New Drug (IND). Such formulation would be treated as

Investigational New Drug (IND) and the general requirement for quality,

safety and efficacy will be same as specified in Schedule Y of D & C Rules,

1945. Independent studies for the carrier, in such cases, may not be required.

3. Conventional/ traditional form of the drug is approved in well regulated

countries and/or India but the nanocarrier system is new and not approved in

the any country. For this category of nanoformulation product, the entire

requirements of safety and efficacy data as specified in Schedule Y for

Investigational New Drug (IND), may not be required. However for

evidence of safety and efficacy of the product should be documented.

4. Conventional/ traditional form of the drug and the nanocarrier system both

are approved in well regulated countries and/or India. It should be subjected

to abbreviated studies.

Note: It is to be noted that the requirements for quality, safety and efficacy

of any nanopharmaceutical should be decided depending upon factors like

physicochemical nature, biological

19
Guidelines on Nanopharmaceuticals

nature, functions, bioavailability and biodistribution, possible interaction

with biological system or exogenously administered medications,

therapeutic indication for which the product is intended to be used, route of

administration, intended duration of therapy, age of the patient, background

data available on the Active Pharmaceutical Ingredient (API) and

nanocarrier, the regulatory status in other countries, etc3.

20
Guidelines on Nanopharmaceuticals

6 Scientific Rationality of making the Nanopharmaceutical preparation

The rationality of making a nanopharmaceutical should be specified with

reference to its added advantage and possible disadvantage in comparison to

conventional/traditional drug. The nanocarriers and its waste disposal may have

adverse impact on the environment and ecosystem. While justifying the

rationality, the known and perceived adverse impact on environment should

also be taken into consideration4.

The following aspects should be specifically addressed for justification of a

nanopharmaceutical:

• Basis of making the claim of improved safety, efficacy, reduction in

toxicity profile, reduction in dose, frequency of administration of the

nanopharmaceutical, improved patient compliance, lower cost or any

other benefit over conventional, traditional drug.

• Addressing any issue arising out of significantly different

pharmacokinetics (PK) and/or pharmacodynamics (PD) than that of the

conventional/ traditional drug.

• Addressing the issue of specific adverse effect/ property of the

conventional/ traditional drug, if any, such as teratogenic potential,

21
Guidelines on Nanopharmaceuticals

Central Nervous System (CNS) side effects, cardiovascular side effects,

QTc prolongation, ophthalmic side effects, etc.

22
Guidelines on Nanopharmaceuticals

7 Specific Considerations for Evaluation of Nanopharmaceuticals in the

context of Schedule Y of Drugs and Cosmetics Rules, 1945

These guidelines have been developed in line with the provisions of

Schedule Y of Drugs and Cosmetics Rules, 1945, with specific requirements for

nanopharmaceuticals wherever considered necessary. While Schedule Y specifies

the general requirements and guidelines for manufacture or import of new drugs or

to undertake clinical trial, this document also provides guidance for specific

requirements of chemical and pharmaceutical information, non clinical data and

clinical data relevant for any product developed based on nanotechnology.General

requirements as specified in Schedule Y will be applicable for any new drug

whether nanotechnology based or not. However, because of inherent complexity

involved in nanotechnology based products, a ‘case by case basis’ approach should

be adopted for evaluating their quality, safety and efficacy.

23
Guidelines on Nanopharmaceuticals

8 Stability Testing of Nanopharmaceutical

The stability testing of nanopharmaceuticals should be done according to the

general requirements specified in Appendix IX of Schedule Y of Drugs and

Cosmetics Rules, 1945.

Stability testing of developmental nanopharmaceuticals must be done

extensively, and systematically. As the drug is loaded in the nanocarrier, the

stability of the drug in an active form should be tested from time to time. It

should focus on functionality, integrity, size of nanoparticles, carrier material

stability, drug stability, degradation products, etc. It should be ensured that the

selected stability storage conditions are relevant for the specific product and

studies are done in intended market packs5. In addition, parameters specific to

nanoparticle-based systems need to be quantified at different time intervals such

as size and size distribution commonly measured using Dynamic Light

Scattering (DLS), surface characterization (zeta potential, functionality, surface

chemistry). In case of surface coating for example with PEG, the PEG layer

thickness should be measured by small-angle X-ray diffraction. The morphology

of the nanoproduct should be determined by microscopy. The residual drug in

the system with reference to initial drug loading and drug encapsulation should

be assessed. Characteristics specific to a subcategory of nanoparticle-based

systems may need to be characterized, for example lamellarity for liposomes,

24
Guidelines on Nanopharmaceuticals

which can be evaluated using cryo-Transmission Electron Microscope (cryo-

TEM). It is advisable to use multiple analytical methods that complement each

other to evaluate the same parameter, for example, DLS, X-ray Diffraction and

cryo-TEM can be used in parallel for the measurement of particle size.

25
Guidelines on Nanopharmaceuticals

9 Animal Pharmacology Data

Knowledge of the activity and toxicities of the free drug, the behaviour of

different delivery systems, and an understanding of the influence of drug release

rate on target and off-target concentrations of bioavailable drug selection of an

appropriate range of nanopharmaceuticals to test. The overall principle of animal

pharmacology should be according to the broad guideline specified in Appendix

IV of Schedule Y of Drugs and Cosmetics Rule, 1945.

To evaluate nanopharmaceutical or nanomedicine efficacy, pre-clinical research

should generate data sets that evaluate the properties of product behavior. Such

properties include- the accumulation of the drug at the disease site, for example

in case of anti cancer product its high accumulation in tumor, intra- tumoural

distribution, and tumoural retention of the system. In addition, the contribution

of the peripheral pharmacokinetics (or circulation) of the nanopharmaceutical

should be assessed. It is likely that for any targeted delivery system, each of

these features may independently contribute to potential efficacy. Based on the

study results, the dominant feature can influence the choice of delivery system

and desired release kinetics. Further, understanding the off-target effects is as

26
Guidelines on Nanopharmaceuticals

important as evaluating efficacy when nanopharmaceutical is being developed.

The pre-clinical testing with an aim to document the translational potential

should provide detailed insight into the key parameters that influence

nanopharmaceutical efficacy.

The informative and translatable data sets should consider to characterize the

disease site specific retention, drug release rates, and drug metabolism. It should

differentiate between bioavailable/released drug and total concentrations of drug

in the site of action (where applicable, for example tumour), plasma, and other

key organs (e.g., liver, kidney, bone marrow, etc.). The data should help to

evaluate how the plasma, off-target tissue, and disease site (target if applicable)

pharmaco- kinetics are affected by repeat dosing. An effort should be made to

separate the evaluation of parmacokinetics/biodistribution from

efficacy/mechanism of action using nanopaharmaceuticals. The preclinical study

should have a clear focus on the end clinical application (such as combination

with standard-of-care) of the nanopharmaceuticals

For Brain targeted nanopharmaceutical special studies should be done to measure

drug concentration in different parts of brain along with API.

27
Guidelines on Nanopharmaceuticals

10 Animal Toxicology Data

Generation of data in the area of animal toxicology for nanopharmaceuticals

should follow the general guidelines as specified in Appendix III, Schedule Y of

Drugs and Cosmetics Rules, 1945.

The toxicology studies should be conducted in the most clinically relevant

animal model6. Toxicology studies should generally be performed in both sexes,

in a rodent and non-rodent species, usually rats and dogs. In certain cases, if

specific animal species are historically more predicative of toxicity for

certain drug classes (for example, primates for predication of complement-

mediated toxicity of phosphorothionate oligonucleotide therapies), it should be

used for study. In this context, it may be mentioned that due to species-specific

target expression, in some cases only primates are relevant for toxicology

studies. There may be situations where there are no nonhuman target-expressing

animals. In such cases, transgenic animals expressing the target or a surrogate

ligand for a similar animal target can be used to characterize toxicity profiles.

For nanoparticles, uptake by the reticulo-endothelial system (RES) has been

demonstrated to be an important modulator of biodistribution. In this context,

the most relevant species for evaluating nanomaterial toxicology or ADME, with

28
Guidelines on Nanopharmaceuticals

regard to Reticulo-Endothelial System (RES) function, is not clear. The studies

suggest that in laboratory animals (rats, mice, guinea pigs, rabbits and dog) and

man, splenic macrophages and liver Kupffer cells are primarily involved in

sequestration of nanoparticles, while in some larger animals (sheep, goat, cat,

and pig) Pulmonary Intravascular Macrophage (PIM) are primarily involved in

trapping/ sequestration.

The dosing regimen and administration route for repeat-dose toxicology studies

are dictated by the intended clinical administration route and regimen, which is

in turn dictated by the pharmacology of the nanopharmaceuticals.

Toxicology studies should also include the intravenous route for

nanoformulations where the primary clinical administration route is not

intravenous, to allow for high exposure comparison. The duration of multi-dose

toxicology study is dependent upon the intended clinical dosing duration. The

number of animals required for toxicology and toxicokinetic studies depends

upon the study length and statistical significance of the result which in turn is

dependent on variation of result. For example, the studies of up to 4 weeks in

duration, 5–10 rats or 3–4 dogs per each sex per dosage group are usually

sufficient.

29
Guidelines on Nanopharmaceuticals

There are some special issues which may be considered for

nanopharmaceuticals. The maximum dose used in preclinical toxicology studies

depends upon several factors, including the toxicity of the nanoformulation and

its solubility. It is usually not reasonable to dose a nanoformulation over several

g/kg, or 50 fold greater than the expected clinical exposure, based on area under

the time–concentration curve (AUC). If toxicity is not observed at these high

doses, then it is not necessary to escalate further. Alternatively, if the drug is

only soluble or stable at mg/mL concentrations in the optimum vehicle (as is

sometimes the case for nanoformulations), then the dose would be limited by

this solubility and by the maximum volume that can be administered to the

animal model by the clinically relevant administration route and dosing regimen.

The lack of toxicity profile characterization, and an inability to identify a

maximum tolerated dose (MTD) and dose limiting toxicities (DLT), either due

to solubility limitations or instability at high concentrations, complicates risk

analysis and the selection of a first-in-man dose. The identification of the toxic

doses is generally not difficult for cytotoxic chemotherapeutic agents. However,

the biologics, on the other hand, which may not demonstrate toxicity in

preclinical models at reasonable doses, are often dosed to pharmacologically

appropriate blood concentration, based on receptor affinity or biomarker

modulation, and not MTD.

30
Guidelines on Nanopharmaceuticals

It is important to include the drug-free (or empty) nanoparticle and free drug as

control groups in toxicology studies, to allow identification of particle-

dependent toxicities and particle-dependent shifts in the encapsulated drug's

toxicity, respectively.

The toxicity studies should comply with the norms of Good Laboratory Practice

(GLP). These studies should be performed by trained and qualified staff using

calibrated and standardized equipment of adequate size and capacity. Studies

should be done as per written protocols with modifications (if any) verifiable

retrospectively. Standard operating procedures (SOPs) should be followed for

all managerial and laboratory tasks related to these

studies. Nanopharmaceuticals (test substances) and test systems (in-vitro or in-

vivo) should be characterized and standardized. All documents belonging to

each study, including its protocol, raw data, draft report, final report, and

histology slides and paraffin tissue blocks should be preserved for a minimum of

5 years after marketing of the nanopharmaceutical.

Toxicokinetic studies of the nanopharmaceutical (generation of ADME data

either as an integral component of the conduct of non-clinical toxicity studies or

in specially designed studies) should be conducted to assess the systemic

exposure achieved in animals and its relationship to dose level and the time

course of the toxicity study. Other objectives of toxicokinetic studies include

31
Guidelines on Nanopharmaceuticals

obtaining data to relate the nanoparticle sequestration based organ exposure

achieved in toxicity studies to toxicological findings and contribute to the

assessment of the relevance of these findings to clinical safety, to support the

choice of species and treatment regimen in nonclinical toxicity studies and to

provide information which, in conjunction with the toxicity findings, contributes

to the design of subsequent non-clinical toxicity studies. Apart from the drug,

the metabolism and excretion of the carrier should be evaluated in cases of

nanopharmaceutical. If toxicity is observed with a nanopharmaceutical, analysis

of results should indicate that the occurrence of toxicity is unrelated or

correlated with API and/or nanocarrier.

An important benefit of some nanopharmaceutical is the ability to formulate a

drug without using dose-limiting toxic excipients present in currently marketed

formulations, thus improving tolerability and enabling administration of more

drug to patients. For example, higher doses of paclitaxel can be administered to

patients using nanoparticle based albumin bound paclitaxel because this

formulation avoids the use of cremophor needed to formulate conventional

paclitaxel formulation. While not considered to be the major focus for many

nanopharmaceutical product development, such solubilization benefits can be

considerably cost-effective. Moreover, by achieving the ‘right safety’ profile,

this approach can make a significant difference to the patients and the clinical

32
Guidelines on Nanopharmaceuticals

outcome, as the maximum tolerated dose of the active agent can be increased by

avoiding the tolerability problems caused by the solubilizing surfactants.

In cases where the nanopharmaceutical does not show any clinically significant

pharmacokinetics difference than API and has no difference in biodistribution,

exemption of some toxicity studies may be given on the basis of ‘case by case

approach’.

33
Guidelines on Nanopharmaceuticals

11 Clinical Trial Data

The general requirements of clinical data and guidelines as specified in Schedule

Y of Drugs and Cosmetics Rules, 1945 apply to the nanopharmaceutical also.

However, nanopharmaceuticals should be demonstrated clinically through

appropriate design, patient selection hypothesis and biomarkers to exploit the

increased permeability and retention of drug. This is due to modification in

pharmacokinetics and tissue distribution of the nanopharmaceutical to improve

its delivery to the site of action. Clinical development of a nanopharmaceutical

using a well characterized drug delivery system will be successful if the

development plan is designed based on clear understanding of parameters

driving the efficacy of the free drug and the in vivo behavior of the delivery

system. Majority of approved nanopharmaceuticals, especially oncology

products, have been designed clinically to exploit the increased permeability and

retention effect. Such effect may minimize the peak concentration of free drug

while increasing the overall bioavailability of the drug, providing prolong

exposure of the drug at the site of action. At times, the development of a

nanopharmaceutical may fail to achieve the clinical end point in terms of lack of

adequate level of efficacy or increased toxicity due to multiple reasons.

Appropriate design of clinical trials based on proper understanding of

accumulation, retention, toxicity and efficacy profile of the agent and correlation

34
Guidelines on Nanopharmaceuticals

between the in vivo behavior and the delivery system is of paramount

importance for successful assessment of clinical profile of the drug. In general,

clinical trials should be conducted in stages. However, depending on the status

of the Active Pharmaceutical Ingredient (API), whether it is an New Chemical

Entity (NCE) or an approved drug molecule and the nano carrier, clinical trial of

appropriate phase may be conducted on a ‘case by case approach’ basis.

The selection of starting dose for clinical trial for nanotechnology based drug is

estimated in a similar fashion to conventional/ traditional drugs. The clinical

starting dose may be determined by dividing the estimated human equivalent

dose (HED) of the rodent, maximum tolerated dose (MTD) by a predetermined

safety factor. The HED for small molecule cancer drugs is typically determined

by surface area (/m2) scaling of the rodent MTD, or the non-rodent MTD if

1/10th the rodent MTD is found to be toxic to the non-rodent species. In

nanopharmaceuticals there may be variation in safety limits.

35
Guidelines on Nanopharmaceuticals

12 Information Required for Evaluation of Nanopharmaceuticals

As already mentioned, the information required for nanopharmaceuticals should be

decided on ‘case by case approach’ basis. However, in general, the following data

should be submitted to the regulatory authority along with the application to

conduct clinical trials and manufacture of nanopharmaceuticals for marketing in

India.

A. Introduction

a. A brief description of the nanopharmaceutical

b. Indication for which it is intended to be used

c. Category to which it belongs (refer to cause 5.1)

d. Justification for developing nanopharmaceutical

B. Chemical and pharmaceutical information

a. Information on the ingredients

i. Drug information (Generic Name, Chemical Name, INN)

ii. Information on nanomaerial used, excipients/ inactive ingredients

iii. Brief description and rationality of the nanopharmaceutical (Refer to

clause 6)

36
Guidelines on Nanopharmaceuticals

b. Physicochemical characterization data of nanopharmaceuticals

i. Individual component (s) (e.g. API, Nanocarrier material)

ii. Chemical name and structure

iii. Empirical formula

iv. Molecular weight

v. Description of the product with

• Size distribution (poly dispersion index),

• Electronmicroscopy (for shape, size and surface texture)

• Surface charge (zeta potential)

• Process of drug loading in the nanocarrier

• Particle size

• pH (in case of liquid formulation)

• Viscosity (in case of liquid formulation)

Note: From the full list of the product’s physico-chemical parameters, some of

them need to be identified as critical quality attributes. They should be listed along

with the product specifications to ensure quality and reproducibility from batch-to-

batch. In addition, a detailed description of the manufacturing process and the

process controls need to be provided.

37
Guidelines on Nanopharmaceuticals

c. Analytical data (nanocarrier/ API/ nanopharmaceutical)

i. Elemental analysis

ii. Mass spectrum

iii. NMR spectra

iv. FT-IR spectra

v. UV spectra

vi. Polymorphic identification

d. Complete monograph specification for the nanopharmaceutical

i. Identification- defined criteria for unique identification of

nanopharmaceutical

ii. Identity/quantification of impurities

iii. Assay

iv. In vitro/ ex vivo release kinetics of the drug/active ingredient (as

applicable)

v. In vitro/ ex vivo degradation kinetics of nanopharmaceutical and void

nanoparticle at various simulated medium

e. Analytical method validations

For nanopharmaceutical

38
Guidelines on Nanopharmaceuticals

• Assay method

• Impurity estimation method

• Residual solvent/other volatile impurities (OVI) estimation method

f. Stability studies of nanopharmaceuticals (for details refer clause 8)

g. Data on nanopharmaceutical formulation

i. Rationale (justification of the nano form)

ii. Dosage form

iii. Route of administration

iv. Composition

v. Details about loading process, chemical bonding/ conjugation between

active ingredient and carrier, surface coating/ modification and

functionalization

vi. In process quality control check

vii. Finished product specification

viii. Excipient compatibility study

ix. Validation of the analytical method

h. Comparative evaluation of innovator product or approved Indian product, if

applicable

39
Guidelines on Nanopharmaceuticals

i. Container and closure system

ii. Assay

iii. Content uniformity

iv. Impurities

v. pH

i. Forced degradation stability evaluation in market intended pack at proposed

storage conditions

j. Packing specifications

k. Process validation

C. Animal pharmacology (for details refer clause 9)

a. Summary

b. Specific pharmacological actions

c. General pharmacological actions

d. Essential, follow-up and supplemental safety pharmacology studies

e. Pharmacokinetics: absorption, distribution; metabolism; excretion(ADME)

D. Animal toxicology (for details refer clause 10)

a. General aspects

40
Guidelines on Nanopharmaceuticals

b. Systemic toxicity studies

c. Male fertility study

d. Female reproduction and developmental toxicity studies

e. Local toxicity

f. Allergenicity/ hypersensitivity

g. Genotoxicity

h. Carcinogenicity

E. Human / Clinical pharmacology (Phase I)

a. Summary

b. Specific pharmacological effects

c. General pharmacological effects

d. Pharmacokinetics- absorption, distribution, metabolism, excretion

e. Pharmacodynamics-early measurement of drug activity

F. Therapeutic exploratory trials (Phase II)

a. Summary

b. Study report(s)

G. Therapeutic confirmatory trials (Phase III)

a. Summary

b. Individual study reports with listing of sites and investigators.

41
Guidelines on Nanopharmaceuticals

H. Special studies

a. Summary

b. Bio-availability / bio-equivalence.

c. Other studies e.g. geriatrics, paediatrics, pregnant or nursing women

I. Regulatory status in other countries

a. Countries where the nanopharmaceutical is

i. Marketed

ii. Approved

iii. Approved as IND

iv. Withdrawn, if any, with reasons

b.Restrictions on use, if any, in countries where marketed /approved

c. Free sale certificate or certificate of analysis, as appropriate

J. Prescribing information

a. Proposed full prescribing information

b. Drafts of labels and cartons

K. Samples and testing protocol/s

Samples of pure drug substance, nanocarrier material and finished product

42
Guidelines on Nanopharmaceuticals

(an equivalent of 50 clinical doses, or more number of clinical doses if

prescribed by the Licensing Authority), with testing protocol/s, full impurity

profile and release specifications.

43
Guidelines on Nanopharmaceuticals

13 Pharmacovigilance of Nanopharmaceuticals

Pharmacovigilance must be carried out throughout the life cycle of the

nanopharmaceutical.

A detailed pharmacovigilance plan along with marketing authorization

application must be submitted by the sponsors.

Pharmacovigilance plan must mention:

• Safety data from clinical development

• All the potential risks of the nanopharmaceutical

• Summary of anticipated risks

• Population at risk and

• Situations not adequately studied

• All the potential drug - drug and drug – food interactions of the

nanopharmaceutical either as a separate document with

pharmacovigilance plan or pharmacovigilance strategies or in the section

referring to safety specifications of the document.

For nanopharmaceuticals of antimicrobials, monitoring of patterns of resistance

will be an important component of pharmacovigilance. Hence, strategies for

monitoring and prevention of the resistance should be mentioned in a separate

44
Guidelines on Nanopharmaceuticals

section of the document. For nanopharmaceuticals of antimicrobial agents, a signal

will be generated if there is an alarming rise in the incidence of resistance to it

for the particular claim proposed.

If any significant safety concerns arise during clinical trials which warrant

studies in special populations such as children, elderly, pregnant women or in

hepatic or renal failure patients, the protocol of such studies should be submitted

along with the pharmacovigilance plan.

Protocols for comparative observational studies (cross sectional/ case control/

cohort), drug utilization study or any targeted clinical evaluation to be conducted

as a part of pharmacovigilance plan should be the part of the document.

45
Guidelines on Nanopharmaceuticals

14 Conclusion

General requirements and guidelines specified for approval of manufacture/import

of any new drug or to undertake clinical trial as specified in the Drugs and

Cosmetics Rules, 1945 especially in Schedule Y and other applicable regulations

apply to nanopharmaceuticals also. However, the requirement of special or

additional tests for safety and efficacy evaluation of a particular

nanopharmaceutical should be decided on a ‘case by case approach’ basis which

will depend upon various factors such as physicochemical and biological nature,

and other aspects including the background data available on the API or

nanocarrier, the regulatory status in other countries, etc. Successful translation of

nanopharmaceuticals from nonclinical proof of concept to clinic is challenging.

Like development of any new drug, it requires effective integration of

nanotechnology with chemistry, lifesciences and medicine. However, because of

complexity in nanotechnology, the system necessitates a ‘case by case approach’

with involvement of varied expertise for successful development of

nanopharmaceuticals.

46
Guidelines on Nanopharmaceuticals

15 References:
1. Fatehi L, Wolf SM, McCullough J et al. Recommendations for Nanomedicine
Human Subjects Research Oversight: An Evolutionary Approach for an Emerging
Field. J Law Med Ethics. 2012 ; 40(4): 716–750.
2. Ragelle H, Danhier F, Preat V et al. Nanoparticle-based drug delivery systems: a
commercial and regulatory outlook as the field matures. Expert Opin Drug Deliv.
2017 Jul;14(7):851-864
3. Navya PN, Daima HK. Rational engineering of physicochemical properties of
nanomaterials for biomedical applications with nanotoxicological perspectives.
Nano Convergence: 2016;3(1):1-14
4. Stern ST, Hall JB, Yu LL et al. Translational considerations for cancer
nanomedicine. ournal of Controlled Release. 2010;146: 164–174
5. Ai J, Biazar E, Jafarpour M et al. Nanotoxicology and nanoparticle safety in
biomedical designs. International Journal of Nanomedicine 2011:6 1117–1127
6. Hare JI, Lammers T, Ashford MB et al. Challenges and strategies in anti-cancer
nanomedicine development: An industry perspective. Advanced Drug Delivery
Reviews. 2017;108: 25–38

16 Glossary

! Biodegradable product

The product that is capable of being broken down or decomposed into

innocuous products/components

! Nonbiodegradable product

The product that is not capable of being broken down or decomposed into

innocuous products/components

! Organic nanomaterials

The nanomaterials that are related to or have been derived from living matter.
47
Guidelines on Nanopharmaceuticals

! Inorganic product

The product that is not derived from or related to living matter.

! Nanoscience

The study of nanomaterials

! Nanotechnology

The application of nanoscience to enable innovations

! Nanocarrier

Nanoparticle/nanomaterial carrying drug or other biomolecules

48

You might also like