You are on page 1of 21

Medicine in Drug Discovery 15 (2022) 100134

Contents lists available at ScienceDirect

Medicine in Drug Discovery


journal homepage: www.elsevier.com/locate/medid

Review Article

Nano-based drug delivery systems: Conventional drug delivery routes, recent


developments and future prospects
Afreen Sultana a, Mina Zare a,b,⇑, Vinoy Thomas c, T.S. Sampath Kumar d, Seeram Ramakrishna a,⇑
a
Center for Nanotechnology and Sustainability, Department of Mechanical Engineering, National University of Singapore, Singapore 117581, Singapore
b
Department of Food and Nutrition, P.O. Box 66, 00014, University of Helsinki, Finland
c
Department of Materials Science and Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
d
Medical Materials Laboratory, Department of Metallurgical and Materials Engineering, Indian Institute of Technology Madras, Chennai 600036, India

A R T I C L E I N F O A B S T R A C T

Keywords: Drug delivery can be defined as the group of approaches a drug or pharmacologically active agent can be trans-
Target drug delivery ported to the target cell to treat disease or health issue. Conventional routes for drug delivery are oral, buccal,
Nanocarriers rectal, subcutaneous, intranasal, intramuscular, intravenous, pulmonary and transdermal. These are the com-
Pharmacokinetics monly used methods for treatment of various medical problems but have certain limitations such as instability,
Pharmacodynamics
risk of displacement, uncontrolled release, side effect such as irritation and pain, slow absorption, enzymatic
Nanomedicine
Smart drug delivery
deterioration and many others. Incorporation of drug into nanocarrier is one of the efficient methods for tar-
geted and sustained delivery of drug. Applications of nanocarriers such as solid nanoparticles, liposomes, den-
drimers, polymeric nanoparticles, polymeric micelles, virus like nanoparticles, carbon nanotube and
mesoporous silica nanoparticles are discussed in this review. To overcome drawbacks of drug delivery, inno-
vative delivery systems are designed usually termed as smart drug delivery systems which include nucleic
acid‐based drug delivery system, cell‐based drug delivery system, self‐nano emulsifying drug delivery system,
self‐micro emulsifying drug delivery system, chemical and physical stimuli‐based drug delivery system, nano-
needles, patches, ultrasound drug delivery and microchip technology. This article focuses of the basic mecha-
nism of drug delivery, pharmacokinetic study, recent innovations and future trends of the drug delivery system.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2. Mechanism of targeted drug release . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.1. Linker cleavage. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.2. Control of carrier . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3. Pharmacokinetics. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3.1. Absorption . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3.2. Distribution . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3.3. Metabolism. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3.4. Elimination. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
4. Pharmacodynamics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
5. Classification of drug delivery systems based on routes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
6. Role of nanocarriers in drug delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
6.1. Solid lipid nanoparticles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
6.2. Liposomes. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
6.3. Dendrimer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
6.4. Polymeric nanoparticles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
6.5. Polymeric micelles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9

⇑ Corresponding authors.
E-mail addresses: zare.mina@yahoo.com (M. Zare), seeram@nus.edu.sg (S. Ramakrishna).

https://doi.org/10.1016/j.medidd.2022.100134
Received 5 February 2022; Revised 13 May 2022; Accepted 15 May 2022
Available online 20 May 2022
2590-0986/© 2022 The Author(s). Published by Elsevier B.V.
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
A. Sultana et al. Medicine in Drug Discovery 15 (2022) 100134

6.6. Virus-based nanoparticles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9


6.7. Carbon nanotubes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
6.8. Mesoporous silica nanoparticles. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
7. Smart drug delivery systems. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
7.1. Nucleic acid-based drug delivery system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
7.2. DNA nanotechnology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
7.3. Spherical nucleic acid . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
7.4. Exosomes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
7.5. Lipid nanoparticle . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
7.6. Aptamer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
7.7. Cell-based drug delivery system. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
7.8. Self-nano emulsifying drug delivery system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
7.9. Oil . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
7.10. Surfactant . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
7.11. Co-solvent. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
7.12. Self-micro emulsifying delivery system. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
7.13. Physical stimuli responsive drug delivery system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
7.14. Chemical stimuli responsive drug delivery system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
7.15. Nanoneedle patches . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
7.16. Ultrasound drug delivery. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
7.17. Microchip technology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
7.18. Nano vaccine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
7.19. Dry vaccine delivery system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
7.20. Bioceramic nanoparticles. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
7.21. Nanogels. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
7.22. Fullerenes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
8. Ethics and regulatory affairs in nano-drug delivery system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
9. Future scope . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
9.1. Drug delivery for future viruses . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
9.2. Safety and efficacy of drug . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
9.3. Symbiotic drug delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
9.4. Gender sensitive drug delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
9.5. Affordable . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
9.6. Greener drug delivery system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
9.7. Intelligent drug delivery system. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
10. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
Author contributions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
Funding . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
Ethical statement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
Conflicts of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16

1. Introduction via constrain mesh. In osmotic pump system, drug is released due to
change in osmotic pressure through hole/ holes in impermeable mem-
Drugs are defined by FDA, in part, as “intended for use in the diag- brane. In erodible material, drug is released when the material gets
nosis, cure, mitigation, treatment, or prevention of disease” and “arti- dissolved1. Self‐emulsifying drug delivery system are composed of
cles (other than food) intended to affect the structure or any function oil, surfactant, co‐surfactant and drug and when diluted with water
of the body of man or other animals. It is expected that the drugs are result in micro or nano emulsion [3]. Implementation techniques are
capable of targeting the disease‐causing cell with an exact therapeutic medium via which drug can be delivered such as microsphere (spher-
concentration in an effective manner. However, in contrast, it is ical particle with diameter in range between 1–1000 μm) [4] and tubu-
observed that release rate, stability and cell‐ and tissue‐specific target- lar vesicles (persistent length is greater than microsphere) [5]. Clinical
ing ability are uncontrolled and cannot be monitored [1]. To overcome application involves the investigation of drug validity as per the clin-
challenges, drug delivery system is designed. Drug delivery system has ical view point [2].
the capability to regulate drug release rate and improves the effective- There are various conventional ways to transport drug to the target
ness of the drug (Fig. 1) [2]. cell, but all these methods have certain limitations. This has led to the
Designing of drug delivery system includes the study of engineering development of smart drug delivery system which is capable of over-
concepts, material design, implementation techniques and clinical coming the shortcoming of conventional methods. This review focuses
application. Engineering concepts focuses on the diffusion, erosion, on the mechanism involved in targeted delivery of the drug, discusses
degradation, shear, swelling, binding kinetics, passive cell uptake, sur- the four important steps involved in movement of drug and the con-
face area and active cell uptake. Material design includes two systems‐ ventional and smart drug delivery systems. This study is impactful
controlled drug delivery system and self‐emulsifying system [2]. Con- for the researchers to understand the basics and future advances in
trolled drug release system is classified as matrix, reservoir, degrad- the drug delivery system.
able material, hydrogel, osmotic pump and erodible material. In
matrix system, drug is released through interconnected pores. In reser- 2. Mechanism of targeted drug release
voir system, drug permeates through the semipermeable membrane. In
degradable material system, the material degradation causes porous Targeted drug delivery system is system in which pharmacokinetic
structure leading to extrusion of drug. Hydrogel system release drug drug is transported to the site of action and prevents the unnecessary

2
A. Sultana et al. Medicine in Drug Discovery 15 (2022) 100134

Fig. 1. Schematic illustration of factors involved in designing of effective drug delivery system. These factors include safety, efficacy, preparation technique and
adsorption.

interaction with other healthy tissue to avoid side effects [6]. Non‐ ers is not triggered by any chemical trigger [11]. Linker cleavage plays
targeted drug delivery such as chemotherapeutic drug used for cancer a vital role in targeted delivery of drug which is further discussed in
treatment leads to undesirable effect on healthy cells. Targeted drug this section. The drug release through the linker cleavages can be per-
delivery improves the uniformity of the drug effect and reduces the formed via various mechanisms such as ester hydrolysis, amide hydrol-
drug dosage [7]. Targeted drug release is a three‐step process: (i) ysis, hydrazone hydrolysis, disulphide exchange, hypoxia activation,
through multivalent receptor–ligand interactions nanocarrier binds mannich base, self‐immolation, photochemistry, azo reaction and
with the receptors of the target cell, (ii) through endocytosis drug thermolysis.
nanocarrier enters into the cell and (iii) in the last step drug release Ester linkers work well at basic pH as well which is useful for con-
takes place. Targeted drug delivery can take place in cytosol and cell trolled release [12]. Amide linker can be used for formation of drug
membrane by interacting with lipid membrane (Fig. 2) [8]. conjugate with nanocarrier. For chemical hydrolysis of amide bond,
Drug release can be performed via two methods: linker cleavage high temperature along with strong acid or base is needed [13].
and control of carrier which are further discussed in this section [8]. Hydrazone bond is stable at 7.4 pH and the optimal pH for cleavage
is equal to or more than five [8]. Disulphide bond cleavage takes place
2.1. Linker cleavage in cytoplasm due to electrochemical reaction or disulfide exchange
reaction and is triggered by thiol group [14–17]. Hypoxic condition
Constituent of fusion proteins which are capable of separating mul- is the one in which oxygen supply is low in tissue and cell such as
tiple domains in a single protein is known as linker [9,10]. Linkers are tumor microenvironments [18,19]. Mannich base acts as a prodrug
of two types cleavable and non‐cleavable. Cleavable linkers easily for the parent drug which helps in enhancing the pharmacokinetics
break bond in the presence of suitable condition. For instance, anti- attributes of the drug [8]. Self immolative linker system does not
body–drug conjugates enter into to the lysosome via endocytosis. Lyso- release drug directly instead involves intervening linker. Once the
some has an environment where pH is low and contains hydrolytic reaction is triggered, triggering moiety is excluded from the linker,
enzymes which promotes the cleavage. Cleavage of non‐cleavable link- leading to activation of free spacer and spontaneously causing drug

3
A. Sultana et al. Medicine in Drug Discovery 15 (2022) 100134

Fig. 2. Schematic illustration of target drug release system in cytosol and cell membrane. It shows that nanoparticles get linked to the receptors, then via
endocytosis enters the cell and ultimately leads to release of drug.

release. It has an advantage over other mechanisms that is it amplifies In a study it was reported that permeability of transdermal drug deliv-
the release frequency by repetitive release of numerous drug units per ery can be improved by using microneedle in the presence of ultra-
reaction cascade8. Photochemistry involves the use of photocleavable sound [32]. Nanostructures are also one of the ways to overcome
linker and this reaction is activated in the presence of light [20,21]. these drawbacks [33]. Nanostructures can be fabricated using electro-
Azo reaction involves cleavage of azo linker via biological mechanism spinning to improve efficacy and avoid loss of effectiveness of active
which can be used to activate anti‐inflammatory group of azo‐linked component [34].
prodrugs of 5‐aminosalicylic acid [22,23]. Thermolysis refers to cleav-
age of chemical bond of drug linker where thermal stimuli act as a trig- 3.2. Distribution
ger [8].
At distribution stage, drug moves from systemic circulation to the
2.2. Control of carrier tissue. Nanocarrier helps in prolonging the blood circulation and pro-
mote target delivery [31] (Su et al., 2019). One of the major challenges
Non‐covalent mechanism can be used for drug loading and release. in drug distribution is blood brain barrier, which can be improved by
It can be performed via encapsulation or interaction with the carrier intranasal administration, transcytosis or direct injection of drug to the
where particle size, shape and geometry have a vital role in regulating central nervous system [35]. For instance, in a study, it was recorded
biological mechanism [24–27]. In encapsulation mechanism, drug is that on exposure to ultrasound, permeability improved, releasing the
loaded into the carrier and the drug is released via diffusion in a con- ultrasmall super‐paramagnetic iron oxide nanoparticles from the
trolled way [8]. microbubles and opened the blood brain barriers [36].

3.3. Metabolism
3. Pharmacokinetics
Metabolism is a process of breaking down drug using enzymes,
The essence of pharmacology is the relationship between the dose which can occur in liver, gastrointestinal tract, kidneys, lungs or skin.
of a drug given to a patient and the resulting change in physiological There are two phases for metabolism of drug, but in some cases both
state (the response to the drug). The absorption, distribution, metabo- the phase occurs simultaneously for complete metabolism [37,38].
lism, and excretion of a molecule define its pharmacokinetics, and Nanocarrier are removed from the body via structural degradation
many of these processes, in turn, are controlled by the physicochemi- [31] (Su et al., 2019). In a study it was reported that immobilization
cal properties of the molecule. Pharmacokinetic helps in understand- of Cytochrome P450 onto electron can eliminate the need of NADPH.
ing these four important processes [28]. Hence, it can enhance the metabolism process by reducing the compli-
cations [39]. Thus, these factors need to be considered while designing
3.1. Absorption of drug [40,41].

Absorption step refers to the movement of drug from the adminis- 3.4. Elimination
tration site to the body circulation system. Absorption of drug to the
systemic circulation can occur via five routes such as transcellular Elimination of drug and metabolites can occur via several routes
absorption, pinocytosis, transport protein mediated absorption, para- but the most significant is via kidney. Elimination via kidney involves
cellular, and endocytosis [29]. Low aqueous solubility and poor per- glomerular filtration, secretion of active tubular and then reabsorbs
meability of drug are the main obstructions in drug absorption [30]. tubular [42]. Excretion can occur via sweat, bile, urine, breast milk
Adsorption of an nanocarrier occurs via internally interacting with and saliva [43]. Elimination of nanocarriers depends on shape, size
organism or permeating through biobarriers [31] (Su et al., 2019). and charge of the nanocarriers [31] (Su et al., 2019).

4
A. Sultana et al. Medicine in Drug Discovery 15 (2022) 100134

Targeted delivery of nano drug can be obtained via intercellular The pharmacokinetics profile of nanocarriers is different in terms of
transport, intracellular transport, controlling particle size, enhanced biodistribution, degradation, and the drug release behaviour and it
permeability and retention, coating with hydrophilic component (un- also affects the pharmacodynamic profile. The nanodrug gets accumu-
hygenic) and conjugating with other materials such as peptide. These lated in target sites through two pathways, one in which the nanocar-
methods facilitate the adsorption, metabolism, distribution and excre- rier drug gets diffused in the target cell and second is in which
tion of drug. It also helps in reducing the cytotoxicity against normal nanocarrier loaded with drug gets distributed to the target site and
cells and enhances the efficacy [44]. releases the drug [46] (Liu et al., 2016).

5. Classification of drug delivery systems based on routes


4. Pharmacodynamics
Drug can be administered through several routs such as oral, buc-
Pharmacodynamics is the process to determine the magnitude and cal, rectal, subcutaneous, intravenous, intranasal, intramuscular, pul-
type of drug responses when it reaches the site of action using quanti- monary and transdermal. These drug delivery systems have some
tative tools. Drug response can be defined as the chemical interaction advantages and limitations as well which are mentioned in the
between drug and binding site. Binding site (its known as receptors if Table 1.
it consists of functional activity) are the sites at which drugs bind to As mentioned in the table, all the drug delivery systems have cer-
macromolecule. Drug binding leads to activation of receptors and tain shortcomings for which various studies are being performed to
influences the intracellular messengers or proteins. overcome such challenges. In a recent study, smart microencapsulation
Receptors are classified into five categories: (i) ligand gated ion system is designed to overcome challenges in oral drug delivery. In this
channels: ligands bind to channel to regulate the flow of ions through research oil in water emulsion solvent evaporation method was used to
the cell membrane (example: nicotinic acetylcholine receptor), (ii) fabricate hollow microparticles (Eudragit S100 polymer) consisting of
voltage gated sodium channel: aqueous pore is formed by agglomera- environmental pH sensitive macrospores [62]. In an investigation,
tion of subunits of receptors through which sodium ions flow, (iii) G mussel inspired mucoadhesive film was developed to attain effective
protein coupled receptor: it converts extracellular stimuli into intracel- attachment to buccal tissue [63]. In a study, mucoadhesive chitosan
lular signals, (iv) Enzyme linked receptors: cytosolic enzyme activity hydrogels were prepared to study the effect on treatment of ulcerative
gets inhibited or activated when ligands bind to such receptors and colitis. It was concluded that catechin‐ chitosan incorporated with sul-
(v) intracellular receptor: ligands has to permeate into the cell to inter- fasalazine is an effective way to treat ulcerative colitis via rectal route
act with these receptors [45]. [64]. Treatment of brain tumor via intravenous drug delivery is chal-

Table 1
List of advantages and disadvantages of various drug delivery systems.

Drug delivery systems Advantages Disadvantages Ref.

Oral drug delivery system Non-requirement of skilled person. Challenging for patients of pediatric, geriatric, and mentally [47,48]
Non-invasive. retarded population.
Cost effective. Difficulty in consumption.
Instability of drug under environmental conditions of
gastrointestinal tract.
Unnecessary accumulation leads to low bioavailability.
Buccal drug delivery Good blood supply and perfusion rate promotes rapid absorption. Residence time. [49–52]
system Improved bioavailability. Some drugs are instable at buccal pH.
Safe from degradation due to environmental conditions of Risk of displacement.
gastrointestinal tract. Drugs which require high dosage cannot be administered.
Lesser dose frequency needed and has small treatment period.
Rectal drug delivery Absence of enzymes helps in avoiding enzymatic degradation. Rectal irritation [53,54]
system Can be administered by patients of pediatric, geriatric, and mentally Absorption can be interrupted by defecation
retarded population.
Effectiveness of localized treatment of problems such as constipation is
enhanced.
Intravenous drug delivery Short latent period. May cause undesirable immune reaction (in case of protein and [55,56]
system Fast achievement of blood concentration required for drug effective peptides).
functioning. May cause thrombophlebitis and tissue necrosis.
May also lead to air embolism.
Subcutaneous drug Sustainable absorption of drug Rate of absorption is slow. [56,57]
delivery system Can be conveniently self-administered May cause pain and irritation at the injection site.
May lead to tissue damage due to accretion of unabsorbed drug.
Intramuscular drug Rapid and uniform absorption. It may cause pain at injection site. [56,58]
delivery system Larger amount of drug can be injected comparatively to subcutaneous Quantity injected depends on the mass of the muscle.
drug delivery method. May cause hematoma.
Accidental rupture of blood vessel may lead to incorporation of
drug into the blood.
Intranasal drug delivery Rapid onset of action. Intolerance in nasal mucosa. [59]
Convenient to use. Poor permeability is observed for hydrophilic agents.
Prevents interaction with gastrointestinal tract.
Pulmonary drug delivery Vascularization and circumvention promote enhanced absorption of Aerodynamic filter inhibits the efficient deposition. [56,60]
system the drug. Mucus lining moves the deposited agents towards the throat.
Inhibits the risk of side effects due to targeted delivery.
Requires lesser dosage
Transdermal drug delivery Prevents deterioration of drug due to gastrointestinal interaction Skin is impermeable to hydrophilic components. [61]
system Convenient to administer May cause enzymatic deterioration.
Incomplete utilization of the drug.

5
A. Sultana et al. Medicine in Drug Discovery 15 (2022) 100134

Fig. 3. Schematic illustration of nanocarriers used in drug delivery which includes solid nanoparticles, liposomes, dendrimers, polymeric nanoparticles, polymeric
micelles, virus like nanoparticles, carbon nanotube and mesoporous silica nanoparticles.

lenging because of blood–brain barrier. Wireless electronic tion, focused ultrasound along with microbubbles has proved to open
doxorubicin‐loaded patch was experimented on nude mice via human blood brain barrier. Results showed that 7.2 µl microbubbles/kg/min
xenograft glioblastoma model. The results showed decrease of tumor or below were able to consistently open blood brain barrier without
recurrence and survival rate was increased [53]. In another investiga- causing any damage [65]. One of the challenge subcutaneous drug

6
A. Sultana et al. Medicine in Drug Discovery 15 (2022) 100134

Table 2 Table 4
Summary of the SLNs for improved drug delivery. List of drugs loaded in dendrimers for effective drug delivery.

Drug loaded Results Ref. Drugs Dendrimers Results Ref.


in SLN
Human Polyamidoamine This model showed the [94]
Carbendazim According to release profile, 30% of Carbendazim was [78] epidermal inhibition of mammosphere
released by 6th day from SLN, suggesting the sustained growth factor formation of MDA-MB-231
release receptor-2 and MDA-MB-231/
(HER2) HER2 + cells. Suggesting
Paclitaxel After 24 hours of treatment IC50 (i.e., 50% inhibitory [79]
selective cancer killer gene
concentration) of Paclitaxel SLN on B16F10 cells was
therapy
recorded as 200 nM and 7.7 mM approximately for
Poly (propylene FTIR report suggest that 5 µg [95]
dacarbazine
imine) maltose- per day of G4-His-Mal
Enrofloxacin SLN loaded enrofloxacin accumulated 27.06–37.71 times [80] histidine (G4-His-Mal) dendrimers decreased the
more efficiently than free enrofloxacin. 0.24 and 0.06 μg/ dendrimers formation of amyloid
mL SLN loaded enrofloxacin was effective in inhibition of aggregates
Salmonella CVCC541
Doxorubicin L-cysteine modified Under acidic condition, drug [96]
Berbamine 25 μM of Berbamine loaded SLN inhibited the invasion of [81] G4.5 dendrimer release from dendrimer was
A549 and MDA-MB-231 cells. After 12 days of treatment, increased to approximately
melanoma tumor volume in C57BL/6 mice model for 16%. It inhibited the
control and Berbamine loaded SLN was recorded above proliferation of HeLa cells in
1500 mm3 and below 1000 mm3 respectively zebrafish
Myricetin 0.8 mg kg−1 of myricetin loaded SLN reduced focal sweat [82] SN-38 L-Lysine dendrimer Three times weekly dose of [97]
by 55% (anticancer 4 mg/kg of SN-38 showed
drug) regression of almost complete
tumor. Maximum weight loss
in treated samples was
Table 3 observed as 3.6%
List of drugs incorporated in liposomes for effective drug delivery.
Doxorubicin CXCR4 targeted At pH 5 faster drug release [98]
Drugs Liposome type Results Ref. (LFC131- dendrimers was observed compared to at
DOX-D4) pH 7.4. It binds to breast
Paclitaxel Chitosan Inhibited 86.4% of tumor [87] cancer cell suggesting the
oligosaccharide effective way for cancer
conjugated Pluronic therapy
P123 polymers
Bupivacaine Unilamellar Sustained drug release was [88]
recorded for 6 days
for efficient insertion of drug‐loaded dissolving microneedles (DMNs)
ZL006 HAIYPRH conjugated Drug loaded in Liposome was [89]
via skin. As per the drug release profile comparison data, DMNs micro-
PEGylated liposomes capable of penetrating into the
brain parenchyma via the lancer are much more effective than DMNs loaded patch even for hairy
endothelial cells and effective in skin. After 2 hours of exposure, DMNs patch has released only 56% of
treating ischemic stroke insulin while microlancer released 92% into the skin. In case of hairy
Paclitaxel Glutamic oligopeptides- HAP binding % showed that [90] skin, same value was observed for DMNs microlancer while for DMNs
RGD peptide (PTX- Paclitaxel (from PTX-Glu6-RGD- patch only 26% of insulin was delivered after 2 hours [70].
Glu6-RGD-Lip) Lip) in metastatic bones was 5–8
times higher than free drug.
Suggesting improved targeting 6. Role of nanocarriers in drug delivery
efficacy
Doxorubicin Thioether The minimum average tumor [91] Nanotechnology is the advanced way to cure cancer, infections and
phosphatidylcholines weight was recorded as 0.78 g by neurological disorders [71]. Drug can be delivered using organic
liposomes samples treated with Doxorubicin nanocarriers and inorganic nanocarriers. Organic nanocarriers include
loaded liposome. In the presence
of 10 mM H2O2, 80% of the drug
solid liquid nanoparticles, liposomes, dendrimers, polymeric nanopar-
was released observed suggesting ticles, polymeric micelles and virus‐based nanoparticles. Inorganic
the improved reactive oxygen nanoparticles include carbon nanotubes and mesoporous silica
species (ROS) triggered release nanoparticles. Fig. 3 represents the schematic representation of vari-
ous nanocarriers used in drug delivery.

delivery is limited volume of drug can be delivered (maximum 3 mL). 6.1. Solid lipid nanoparticles
To overcome which high concentration injectables of more than
100 mg/mL need to be designed and studied [66]. Zhang and few Size of solid lipid nanoparticles (SLNs) ranges between
others performed a study to compare the efficacy of Mecobalamin 50–1000 nm which is prepared by melting solid lipids in water and
intramuscular injections vs oral tablets for treating diabetes peripheral emulsifier is added to form stabilized solution [72,73]. Heat liable
neuropathy. It was concluded that Mecobalamin intramuscular injec- drugs, drugs with poor physicochemical compatibility and low phar-
tion is more effective in treating diabetes peripheral neuropathy com- macokinetic profile can be delivered using SLNs [74,75]. SLNs are cap-
pared to oral tablet [67]. One of the challenges in intranasal drug able of eliminating various complications in targeted and sustained
delivery is observed as poor permeability for polar molecules. To over- drug delivery some. In a study, 5‐Fluorouracil was incorporated into
come this issue, in a research hypergravity was found to be an effective SLN via high pressure homogenization with 81% of entrapment effi-
way of improving nasal drug delivery efficiency [68]. In an investiga- ciency. Results showed that 5‐Fluorouracil SLN had 3.6 times higher
tion, rifampin was loaded into mesoporous silica nanoparticle to attain area‐under plasma concentration–time curve compared to5‐
efficient pulmonary drug delivery. Drug release data showed that, after Fluorouracil. According to tumor efficacy study, mean tumor volume
24 hours 95% of drug was released by drug‐loaded mesoporous silica for control, 5‐Fluorouracil and 5‐Fluorouracil SLN (20 mg/kg) was
nanoparticles [69]. In research, patchless microlancer was designed recorded as 375 mm3, 300 mm3 and 200 mm3 respectively. Hence,

7
A. Sultana et al. Medicine in Drug Discovery 15 (2022) 100134

Table 5
List of drugs incorporated in polymeric nanoparticles for effective drug delivery.

Drugs Type of polymeric nanoparticle Results Ref.

Hydrophobic curcumin Chitosan polymeric nanoparticle Sustained release for 3 hours from the polymeric nanoparticle was observed. Highest [103]
inhibition zone for Pseudomonas aeruginosa was recorded as 3.2 cm
Doxorubicin-triphenylphosphine Polylactic-co-glycolic acid (PLGA) After 12 hours of treatment, MCF-7 cell, at pH 6.5 caused 74% of cell apoptosis. Tumor [104]
wrapped with bovine serum albumin volume was reduced from 26 mm3 to 23 mm3.
Curcumin Poly(lactic-co-glycolic acid)-poly Drug loaded polymeric nanoparticle have 14 fold higher diffusivity in brain parenchyma [105]
(ethylene glycol) (PLGA-PEG) compared to PLGA. Median area loss for drug loaded PLGA-PEG was recorded as 12.3%
Ofloxacin Polycaprolactone Sustained release of drug for six hours was recorded [106]

5‐Fluorouracil SLN is capable of inhibiting HCT‐116 subcutaneous it was observed that efficacy, distribution and concentration of drug at
tumor in mouse [76]. The bioavailability of oral drug administration targeted site can be enhanced by using thermosensitive liposomes
can be improved by encapsulating Amphotericin B and Paromomycin which releases drug on increasing the temperature of tumor via ultra-
in SLN via emulsion solvent evaporation. Entrapment efficiency of sound [86]. Several ongoing researches are found to improve the effi-
Amphotericin B and Paromomycin was recorded as 90% and 96% cacy of drug delivery via liposomes among which some are mentioned
respectively. According to in vitro anti‐leishmanial study, 1 µg/ml of in the Table 3.
modified dual drug loaded solid lipid nanoparticles showed 96.22%
of inhibition against the growth of Leishmania donovani [77]. Table 2.
6.3. Dendrimer
Represents the some of the SLNs studied for improved drug delivery.
Dendrimer consists of several arms originating from core which are
6.2. Liposomes prepared using sugar, nucleotides and amino acids [83]. Dendrimer
are effective method for treating cancer as it depletes bioavailable cop-
Liposomes are spherical vesicle composed of lipid bilayer enclosing per from tumours. Conventional method for synthesis of dendrimer is a
aqueous core which are capable of carrying bioactive agent such as tedious process, therefore effective method was designed to produce
drug [83]. Manufacturing of liposome, loading of drug and removal
of undesirable components can be performed in the continuous process
Table 7
via two microfluidic devices [84]. Although liposomes are effective
List of drugs incorporated in virus-based nanoparticles for effective drug
drug delivery system but lack in on‐demand content release. For this
delivery.
reason, X‐ray‐triggerable liposomes are designed incorporated with
verteporfin and gold nanoparticles. In this investigation it was Drugs Type of virus like Results Ref.
nanoparticles
reported that 6 MeV of X‐ ray triggers the photosensitive vertopoerfin
to produce singlet oxygen and this causes destabilization of liposomal DOX Truncated hepatitis B Within 24 hours almost [120]
membrane causing release of cargos [85]. Similarly, in another report virus core antigen the entire drug was
(tHBcAg) loaded with released. And at pH 5.4,
drug and conjugated more efficient drug
with folic acid release was observed.
Table 6
The IC50 DOX values of
List of drugs incorporated in polymeric micelles for effective drug delivery. virus like nanoparticle
Drugs Copolymer Results Ref. for HT29, Caco-2 and
CCD-112 cells were
Paclitaxel Poly(l-histidine) Minimum increase tumor [110] recorded as 0.14, 2.02
volume (95 mm3) was and 8.10 μM receptively
observed in shielded
Streptokinase Tobacco mosaic virus It enhanced thrombolysis [121]
polymeric micelles loaded
with drug DOX MS2 spheres, tobacco Survival days of mouse [122]
mosaic virus disks and bearing small tumor was
Doxorubicine Diselenide-crosslinked Drug loaded micelles [111]
nanophage filamentous recorded around
micelles (DCM) delivered 3.73-fold higher
rods 40 days days for mouse
quantity of drug compared to
treated with tobacco
free drug in tumor bearing
mosaic virus disks.
mice. Tumor volume for free
drug treated and DOX-DCM Monomethyl Plant virus Potato virus X Reduction in off target [123]
treated samples were 680.86 auristatin drug delivery is
and 210.19 mm3 respectively observed. Median
survival of drug loaded
Doxorubicine Poly(ethylene glycol) At pH 5 drug release was [112]
virus like nanoparticle
observed to be increased two
treated sample was
folds. But the highest
recorded as 54 days
releasewa recorded on
addition of Glutathione Maleimide- Physalis mottle virus More than 83% of [124]
functionalized platinum was released at
Doxorubicine Biotin-Poly(ethylene Cell viability of COS7 cells in [113]
cisplatin pH 5.2 within 48 hours.
glycol)-blocked-poly(L- the presence of micelles was
prodrug More than 90% of mice
lysine) grafted with recorded as 85% and it didn’t
containing Pt (having tumor), treated
matrix possess cytotoxicity
(IV) with drug loaded virus
metalloproteinases suggesting the targeted
like nanoparticles
delivery
survived till 38 days
IR-780 DSPE-PEG2000 Analysis showed; lymphatic [114] while, all control samples
metastasis suppression by were succumbed to
88.2% disease by 28 days

8
A. Sultana et al. Medicine in Drug Discovery 15 (2022) 100134

Table 8
List of drugs incorporated in carbon nanotube for effective drug delivery.

Drugs loaded in CNTs Results Ref.

Silibinin 3T3 cell viability ranged between 69 and 98%. Chitosan coated muliti-walled CNTs showed the highest thermal stability. Sustained release of [133]
drug was observed for 2500 minutes
Doxorubicin In the presence of cathepsin B and at pH 6.5, 85% of the drug was released within 48 hours. Efficient reduction of HCT116 tumor cells by carbon [134]
hydrochloride nanotube was observed
Droxidopa Drug was loaded into the carbon nanotubes efficiently and absorption energy was recorded as −0.714 eV. Bioavailability was enhanced. [135]
Methotrexate On increasing the CNTs, burst release rate decreased. Addition of 0.5% of CNT decreased cell proliferation. Combination of CNT (0.1%) with [136]
hydrogel improved antitumor effect
Formononetin IC50 of Formononetin loaded CNTs for MCF-7 cells and Hela cells was recorded as 17.9% and 21.7% respectively, suggesting the improved [137]
cytotoxicity. Higher release rate was observed at Ph 7.4 than at 5.3 Ph

poly(acyl thiourea) and polythiourea dendrimers. This method [109]. Table 6 represents the list of various polymeric micelles used
involves isothiocyanate–amine coupling and thiol‐methacrylate for effective drug delivery.
Michael addition reaction to produce one dendrimer generation within
four hours [92]. Several studies have been performed on utilizing the 6.6. Virus-based nanoparticles
beneficial effect of dendrimer in drug delivery. Such as in an investiga-
tion, dendrimer incorporated with N‐acetyl‐L‐cysteine (DNAC) was Virus‐based nanoparticles are strong protein cages prepared by self‐
studied for controlling preterm birth risk. DNAS was capable in reduc- assembly which can encapsulate protein, peptide, nucleic acid and
ing the recruitment of CD3 + T‐ cell to the placenta. According to drug [72,115,116]. These protein cages are much small in size which
analysis, 10 mg/kg of DNAC showed more potent results in decreasing leads to difficulty in functionalizing with virus‐binding moieties. To
preterm birth rate than 100 mg/kg of N‐acetyl‐L‐cysteine [65]. Den- overcome this problem, DNA origami triangular tiles was designed
drimers are effective gene delivery system but lacks the application with the ability of self‐assembly into icosahedral shells [117]. Virus‐
due to cytotoxicity. One of the methods to overcome this shortcoming based nanoparticles plays a vital role in targeted delivery of hydropho-
is PEGylation of dendrimers. For instance in an investigation, PEGy- bic drug to tumor cells for therapy. For instance, virus‐like particles of
lated dendrimers showed lesser cytotoxicity on B16F10‐Luc cells com- Flock House Virus was used to design a viral vector. In this system
pared to unmodified dendrimers [93]. Table 4 represents the list of 2 kDa PEG spacer arm protected the nanoparticle from immune reac-
various dendrimers used for effective drug delivery. tion and tLyP‐1 promotes the delivery of drug to the site of action

6.4. Polymeric nanoparticles


Table 9
List of drugs incorporated in mesoporous silica nanoparticle for effective drug
Polymeric nanoparticles are colloidal particles composed of
delivery.
biodegradable polymer which can exist in two forms nanocapsule
and nanosphere. Nanocapsule is prepared by dissolving or dispersing Drugs Nanocarrier design Results Ref.
the drug in liquid core (oil or water) which is encapsulated with poly- Arsenic MSNs coated with PTX was released at pH 6.5 [143]
meric membrane. Nanosphere is fabricated by entrapping drug in poly- trioxide polyacrylic acid and pH- and ATO was released at
meric matrix [99]. Preparation of nanocapsule is a complex process, to (ATO) and sensitive lipid pH 5 effectively
paclitaxel
combat this challenge one‐step self‐assembly strategy was developed.
(PTX)
In a study it was used for encapsulating doxorubicin hydrochloride
ML336 Lipid coated MSNs Dose of 0.11 g LC-MSNs/ [144]
and NaYF4:Yb,Er@NaGdF4 in poly (DL‐lactic‐co‐glycolic acid).
kg/day for four days didn’t
Release profile was dependent on pH, at pH 5, 35.7 wt% and at pH show any toxicity. After 24
7.4, 14.58 wt% of drug was released [100]. In a study, docetaxel‐ hours of treatment, viral
loaded hyaluronic acid nanocapsules were prepared and release profile load was decreased by 4
suggested sustained release of docetaxel for 24 hours [101]. Das and orders of magnitude

colleagues fabricated a poly‐L‐glutamic acid embedded mesoporous DOX MSNs were grafted with After 72 hours, 70% of the [145]
bioactive glass nanosphere. Daunomycin was loaded into the nano- Triphenylphosphine, DOX drug was released and on
was incorporated and addition hyaluronidase,
sphere and results showed higher release at 5.5 pH than at 7.4 pH.
capped with hyaluronic release was increased.
Thus, it can be used for localized drug delivery [102]. Table 5 repre- acid More than 80% of cell
sents the list of various polymeric nanoparticles used for effective drug viability was observed at
delivery. concentration 25 μg/mL
Clofazimine Drug is chaperone by Release efficiency of drug [146]
acetophenone was recorded as 47.2%.
6.5. Polymeric micelles Antibacterial study showed
that 22 µg/mL drug loaded
Polymeric micelles are spherical shell which self assembles using clofazimine was capable of
amphiphilic di‐ or tri‐block copolymers in aqueous media [107]. In a killing 90% of
Mycobacterium
study, controlled radical polymerization was used to prepare amphi- tuberculosis
philic diblock copolymer micelles having hydrophobic core and hydro-
Silver MSNs is incorporated with Minimum inhibition [145]
philic shell. It was observed that on increasing the degree of silver boron nanoparticles concentration of MSNs-
polymerization, release rate of doxorubicin for nine hours decreased (MSNs-AgBrNPs) AgBr NPs to inhibit
from 11.3% to 6.12% [108]. In an investigation, camptothecin was Mycobacterium
incorporated into core of the polymeric micelles and AS1411 aptamer tuberculosis growth was
recorded as 31.25 μg/mL
was covalently attached. Results showed 93% of inhibition of tumor

9
A. Sultana et al. Medicine in Drug Discovery 15 (2022) 100134

Fig. 4. Schematic illustration of DNA nanostructure which acts as the one of the effect methods for drug delivery and is further classified as DNA tile, DNA origami
and DNA wireframe.

[118]. Virus‐based nanoparticles acts as a promising scalable nanopar- size consisting of different pore sizes [138,139]. Sol‐gel process is
ticle platform technology for imaging or detection of disease [119]. the commonly used method to synthesize MSNs which involves
Such drug delivery can be used to prevent off target delivery and hydrolysis and condensation of silicon alkoxide precursors [140].
achieve sustained release of drug some of them are mentioned in the Delivery of drug before it reaches target is one of the commonly faced
Table 7. issue, but using MSNs it can be overcome as the exposure to internal or
external stimuli would trigger the pore opening and permit drug
6.7. Carbon nanotubes release [141]. In an investigation, anti‐carbonic anhydrase IX antibody
was bonded on the surface of MSNs and doxorubicin hydrochloric was
Carbon nanotubes (CNTs) are prepared by folding grapheme sheet loaded. According to in vivo therapeutic efficacy, drug loaded in MSNs
into tube like structure whose diameter is nanoscale and length is showed the least tumor growth at 11th day of treatment. The inhibit-
thousand times the diameter [125,126]. CNTs are flexible and possess ing concentration 50% values for 24 h for drug loaded in MSNs was
electrical conductivity and biocompatibility [127]. It can be single recorded as 1.1 μg/mL. It was also observed that addition of 10 mM
walled or multiple walled, such as for DOX has stronger bond with glutathione, increased release of doxorubicin at pH 5 was from 28%
multiple walled CNTs and facilitate better drug release from single to 80%, suggesting redox‐responsive release [142]. Such drug delivery
walled CNTs [128]. In a study, Magnetospirillum magneticum bacteria can be used to design effective drug delivery, some of them are men-
were used to fabricate carbon nanotubes. Capacitance of this scaffold tioned in the Table 9.
was improved to six times higher than the pure carbon nanotubes Nanocarrier drug delivery helps in improving bioavailability,
[129]. Dehaghani and collegues encapsulated Isatin in a single walled enhances permeability, reduces drug dosage, prevents degradation,
carbon nanotube. Results showed that eleven molecules of Isatin was active and passive targeting, reduces the risk of side effects due to
easily absorbed in 30 Å long single walled carbon nanotube and aggregation and speed up the curing of disease [146,147]. Apart from
remained encapsulated till the end of molecular dynamic simulation various advantages of nanocarrier, its application is limited due to tox-
[130]. Hence, carbon nanotubes are capable of holding drug to obtain icity, difficulty in scaleup process and low drug loading efficacy [148].
targeted drug delivery. In an investigation, Doxorubicin was loaded in
N‐isopropyl acrylamide Carbon nanotube. It was observed that poly-
mer of short chain length has better stability, interaction and release 7. Smart drug delivery systems
of doxorubicin [131]. In another research, doxorubicin (loaded with
folic acid) was incorporated into the carbon nanotube and blended Although conventional drug delivery systems are widely used tech-
with silk protein to develop injectable hydrogel system. Results niques for treatment of health issues but certain limitations have led
showed sustained release of doxorubicin for 14 days [132]. Such the demand for advanced version for delivery of drug known as smart
nanocarrier can be used in design of system capable of reducing sys- drug delivery systems. Smart drug delivery system can be defined as
temic side effects. Such drug delivery can be used to prevent off target system which has controlled release rate and provides spatiotemporal
delivery and achieve efficient drug some of them are mentioned in the resolution. Drug release in such systems takes place as a response to
Table 8. exogenous stimuli such as electric field, ultrasound and magnetic field,
and endogenous stimuli such as enzyme, pH and retention effect
6.8. Mesoporous silica nanoparticles [149]. This review focuses on the impactful smart drug delivery sys-
tems such as nucleic acid, cell‐based, self‐nano emulsifying, self‐
Mesoporous silica nanoparticles (MSNs) are silica material with micro emulsifying drug delivery system, physical, chemical stimuli‐
structure similar to honeycomb and it can be synthesized in varying based drug delivery system, nanoneedle patches, ultrasound drug

10
A. Sultana et al. Medicine in Drug Discovery 15 (2022) 100134

Fig. 5. Schematic representation of formation of aptamer from RNA or DNA which fold into three-dimensional conformation and then binding to the target
resulting in target aptamer complex formation.

Fig. 6. Schematic representation for cell-based drug delivery system which is designed as an inspiration from erythrocytes, platelets, macrophages, leucocytes
(monocytes and neutrophils), T cells, stem cell and bacterium.

delivery system, microchip technology, nano vaccine, dry vaccine drug canonical nucleic acid can be used for diverse structural designing of
delivery system and bioceramic nanoparticles. DNA nanostructures due to its high degree of conformational polymor-
phism [153]. Recent development has led to the exploration of three
pillars of nucleic acid‐based drug delivery system which are DNA nan-
7.1. Nucleic acid-based drug delivery system otechnology, spherical nucleic acids, and aptamers [151].

Nucleic acids are negatively charged, hydrophilic polymer which


are impotent of permeating through the cell, enzymatically unstable, 7.2. DNA nanotechnology
may lead to undesirable biological responses and hence, broadly not
considered as a good option to be used for drug delivery [150,151]. DNA is a nanoscale material which has the ability to carry genetic
But the modified nucleic acid has proved the ability to activate the information. DNA nanotechnology is an excellent method for drug
innate immune system at low concentration [152]. For instance, non- delivery but has the limited use due to its susceptibility to nuclease

11
A. Sultana et al. Medicine in Drug Discovery 15 (2022) 100134

[154]. Advantages of DNA nanotechnology include its precise defined recent study, DNA nanoscale precision‐guided missile was designed
particle architecture, stimuli‐responsiveness and absence of intrinsic which consist of two parts: warhead (rod‐like DNA nanostructure
toxicity. DNA nanotechnology can be further classified as tile‐based which acts as a drug carrier) and a guidance/control (aptamer‐based
assembly, DNA origami, and DNA wireframes (Fig. 4) [151]. logic gate). This DNA nanostructure was capable of specific recogni-
Tile‐based assembly is composed of double DNA duplexes with tion and targeted delivery of doxorubicin for cancer therapy [178].
crossovers [155]. Thermal annealing process is used to fabricate
DNA origami structure, in which staple strand is amalgamated with 7.7. Cell-based drug delivery system
numerous scaffold segments to develop double helical structure
[156]. In a study, DNA origami is incorporated with 2‐[3‐(1,3‐ For designing of cell‐based drug delivery system, erythrocytes, pla-
dicarboxy propyl)‐ureido] pentanedioic acid and then loaded with telets, macrophages, leucocytes (monocytes and neutrophils), T cells,
Doxorubicin via intercalation. Results showed that DNA origami stem cell and bacterium act as inspiration because of their unique
helped in selectively delivering Doxorubicin to the prostate‐specific transportation mechanism (Fig. 6) [179].
membrane antigen cancer cell line LNCaP [157]. Designing of DNA Erythrocytes are red blood cells which due to their special structure
wireframe structure involves three steps: graphical representation and circulating attribute are considered as the efficient source of drug
(2D or 3D) of target structure, network rendering of nodes (vertices delivery system [180]. Platelets are cell whose maximum portion of
of arms) and edges (variable length), separation of edges and sequence cytoplasm is composed of thrombosthenin and nucleus is absent. Pla-
production [158]. In a study, wireframe DNA origami with honeycomb telets have a vital role in hemostasis, whenever there is any injury pla-
edges were designed and it was concluded that structural fidelity is telet acts as source of repairement via four step mechanism: adhesion,
improved [159]. activation, aggregation, and wound repair. Monocytes are leucocytes
with diameter in the range of 50–80 μm, unilobar nuclei and granu-
7.3. Spherical nucleic acid lated cytoplasm and have the ability to migrate into the infected site
by following inflammatory signals. Macrophages are cells with the
Spherical nucleic acids are compact structure composed of DNA diameter of around 21 μm and have the ability to engulf via phagocy-
layers positioned vertically on the center core (silica, quantum dots, tosis and degrade foreign substances. Macrophages have an important
liposomes, polymers, or even empty cavity) [160–165]. In this system, role in innate immunity and adaptive immunity. Neutrophils are gen-
nucleic payloads and co‐delivery of drug occur in synchronized man- erated by the stem cells in bone marrow and belong to polymorphonu-
ner which has beneficial effect on enhancement of stability and solu- clear cell family. Neutrophils play a vital role in innate immunity and
bility, allows delivery without carrier and improved drug loading are well known due to their ability of deformability and chemotaxis. T
[151]. cells are those which mature in the thymus and further divide into sub-
sets. A T cell recognizes epitopes of foreign substance and leads to cell
7.4. Exosomes death. It has the ability of releasing cytokines and enhances immune
system by promoting generation of antibodies via B cells [179]. Stem
Exosomes are extracellular vesicles composed of proteins, mRNAs, cells are unspecialized cells which can differentiate into any cell [181].
lipids and miRNAs. Exosomes which is lipid molecules (30–100 nm In a study it was observed that stem cells are capable of targeting
wide) used for transportation of smaller molecules [166]. Earlier, exo- tumor foci [182]. Bacteria have the ability to locomote along the mole-
somes were considered as cellular waste but research has proved its cules providing signals and this attribute can be used to carry drug to
application in intracellular connection which has effect on functioning specific area [179].
and activity of recipient cell [167,168]. Formation of exosomes For instance, in an investigation, macrophage was used as a pul-
includes four basic steps budding, invagination, formation of multi- monary drug delivery system. Doxorubicin was loaded into cellular
vesicular bodies and secretion of multiple vesicle structures [169]. vectors via electroporation and passively for comparison. In release
Exosomes play a vital role in pathogenesis and progression of health study it was found that, 80% of payloads were released within 2 hours
ailments such as cancer. for passively loaded samples and within 6 hours for electroporation
loading [183].
7.5. Lipid nanoparticle

Lipid nanoparticle are non‐viral gene delivery system which can 7.8. Self-nano emulsifying drug delivery system
deliver siRNA, mRNA, DNA, or gene‐editing complexes [170]. These
nanoparticles are useful in transportation of hydrophobic and hydro- Self‐nano emulsifying drug delivery system (SNEDDS) refers to a
philic molecules. It reduces the toxicity, controlled release and blend, composed of oils, surfactants and co‐solvent which when agi-
increase in drug action [171]. Lipid nanoparticles are profound deliv- tated in aqueous condition results in oil in water nano emulsions
ery system for anticancer drug and as well as environmentally friendly [184–186]. The resulting nano emulsion droplet ranges between
[172]. 20–200 nm and is used for efficient movement of active component
across the intestinal membrane [187]. Fig. 7 represents the compo-
7.6. Aptamer nents of SNEDDS and formation of nano emulsion.
The three major components of SNEDDS are further discussed
Aptamers can be defined as a single stranded oligonucleotide which below:
have the ability to bind to target cells through their 3D structure
[173,174]. Aptamer conjugates are considered as the most effective 7.9. Oil
nucleic acid‐based drug delivery system [151]. Nucleic acid can be
prepared using SELEX (Systematic Evolution of Ligands by Exponential Oil considered for SNEDDS is usually medium to long chain triglyc-
enrichment), which involves the separation of initial nucleic acid bin- erides and should possess good solubility for drug as it facilitates drug
ders from oligonucleotide library, followed by amplification via poly- absorption and formulation‐loading capacity [188]. In contrary, a
merase chain reaction [175] (Fig. 5). study reported that low solubility of drug in oil has better absorption
Aptamers have lesser toxicity, can be easily modified depending on property [189]. Some of the usually considered oils are such as poly-
the requirement, easily bind to tumor biomarkers and inexpensive ethylene glycol, hydrogenated castor oil, sorban esters, glyceryl mono-
(production cost) when compared to antibiotics [176,177]. In an linoleate and triglycerides of capric/caprylic acids [190].

12
A. Sultana et al. Medicine in Drug Discovery 15 (2022) 100134

7.12. Self-micro emulsifying delivery system

Self‐micro emulsifying drug delivery systems (SMEDDS) are com-


posed of oil, water, surfactant and co‐surfactants or co‐solvents which
results in isotropic microemulsion. This blend can be incorporated into
capsule and injested and when it contacts with the gastrointestinal
tract it results in oil in water microemulsion [197]. Major components
of SMEDDS are drug, oil, surfactant and co‐surfactant. Drug used
should not have first pass metabolism and minimal dose is considered.
Medium chain triglyceride moves directly to the circulatory system,
and usually considered for lipid‐based drug due to rapid solubility
and ability to prevent oxidation. Surfactant used should have HLB
value more than 12 to obtain effective emulsification. Medium chain
alcohols are usually considered co‐surfactant. Co‐surfactants are help-
ful in reducing interfacial tension to negative value [196].
One of the examples of SMEDDS includes a study in which oral
bioavailability of cathepsin K inhibitor (HL235) was improved. It
was designed using 5.0% of Capmul MCM EP, 20.0% of Carbitol and
75.0% of Tween 20. The size of the droplet obtain via microemulsion
was 10.7 mm. And the results showed that bioavailability was
improved to 3.22‐fold when compared to control [198]. Some of the
advantage and disadvantages are mentioned in the Table 11.
Fig. 7. Schematic illustration of the four major components of SNEDDS which
are surfactant, co-surfactant, drug and oil. When dissolved in water it results in 7.13. Physical stimuli responsive drug delivery system
formation of nano-emulsion.

Two widely studied physical stimuli responsive drug delivery sys-


7.10. Surfactant tems are thermo and light responsive drug delivery systems. In thermo
responsive drug delivery system, temperature plays a critical role, drug
Surfactants are the substance which reduces surface tension to sta- release depends on temperature change [199]. For instance, in a study
bilize the nanoemulsion. Surfactants can be classified based on charge poly(Nisopropylacrylamide) was used to encapsulate antibacterial
(ionic and non‐ionic) and hydrophilic‐lipophilic balance (HLB) (lipo- peptide of G(IIKK)3I‐NH2 at lower temperature. On increasing the
philic and hydrophilic) [191]. Surfactants which are non‐ionic and temperature above 33 ℃ resulted in release of drug [200]. In light
have hydrophilic‐lipophilic balance more than 12 are considered as responsive drug delivery system, drug release is triggered by the light.
the best option for SNEDDS because they enable unconstrained nano For instance, copolymers of N‐Isopropylacrylamide and N‐
emulsification mechanism [190]. (hydroxymethyl) acrylamide were loaded with sodium copper chloro-
phyllin. On exposure to green laser light for 2 minutes, controlled drug
release was attained to kill cancer cell via 5‐FU loaded nanogel [201].
7.11. Co-solvent
7.14. Chemical stimuli responsive drug delivery system
Co‐solvent or co‐surfactant is required because surfactant alone
cannot always facilitate nano‐emulsification and it helps in attaining Chemical stimuli responsive drug delivery systems are dependent
stability and homogeneity [192]. Co‐solvent and surfactant weight on pH, redox potential and enzyme response. pH responsive drug
ratio has an important role in designing efficient SNEDDS. The mini- delivery system is based on pH‐controlled release of drug. For exam-
mum quantity of co‐solvent is considered to prevent drug precipitation ple, chitosan nanoparticles were incorporated with Tamoxifen can be
which is usually caused due to polarity of co‐solvent which leads to used for targeted drug delivery for tumor treatment. It was observed
migration towards water [193]. that Tamozifen released more efficiently at 4 and 6 pH rather than 7
In an investigation, andrographolide was loaded into SNEDDS to pH+. Change in redox potential also acts as trigger for controlled
study its effect on oral dissolution and bioavailability. The results and targeted drug release. For instance, concentration of glutathione
showed 1.2 folds increase in the AUC (area under the curve) and of cancer cell (2–10 mM) is ten times higher than in normal cells which
1.26 folds increase in Cmax (maximum concentration) for SNEDDS sam- can act as potential gradient for drug release [202]. Enzyme such as
ples when compared to plain andrographolide. Hence, it was con- Cathepsin B (lysosomal protease) can also be used for provoking the
cluded that SNEDDS is effective means for enhancing dissolution and drug release. In an investigation it was observed that polyethylene
bioavailability [194]. Some of the advantage and disadvantages are
mentioned in the Table 10.
Table 11
List of some of the advantages and disadvantages of self-micro emulsifying drug
Table 10 delivery system.
List of some of the advantages and disadvantages of self-nano emulsifying drug
delivery system. Advantages/Disadvantages Ref.

Advantages/Disadvantages Ref. Advantages Improved bioavailability [196]


Thermodynamically stable
Advantages Protects sensitive drug components [195] Manufacturing of SMEDDS is easy
Improved bioavailability and reduces dose intake Food does not interfere with drug absorption
Rapid distribution throughout gastrointestinal tract and Disadvantages In vivo drug precipitation is observed
reduces irritation in gut due to frequent drug intake Handling complication while formulation
Disadvantages It is thermodynamically unstable [196] It has bounded lymphatic uptake
Causes oxidation of unsaturated fatty acid

13
A. Sultana et al. Medicine in Drug Discovery 15 (2022) 100134

glycol‐Gly‐Leu‐Phe‐Gly‐Lys(C16)2 loaded with Doxorubicin resulted microchip was capable of double and serial transfection of the targeted
in controlled drug released due to degradation by capthepsin B and cells [215].
was effective in inhibition of Hep G2 cells viability by 60% [203].
7.18. Nano vaccine
7.15. Nanoneedle patches
Nano‐vaccinology can be defined as science of nanoscale particles
Preparation of Silicon nanoneedle was performed by treating the which has opened an entrance to boundless hopes for treatment of var-
silicon wafer with buffered oxide etch and followed by fabrication of ious diseases [216]. Nanoscale vaccine is designed to achieve sustain-
silicon pillars via photolithography patterning and deep reactive ion able release, improved immunogenicity, improved antigen stability,
etching process. The passivation layer of (CxFy)n polymer is deposited flexible physical characteristics and reduced immune‐toxicity [217].
on the silicon pillar surface under radio frequency power of 800 W. Nanoscale technology is not so common among the present population
Size of silicon pillars is reduced to nanometer by immersing the silicon which makes it difficult to accept and leads to unwanted conspiracies
wafer in potassium hydroxide solution. These silicon nanopillars are such as presence of nanorobotic chips in the vaccine. For covid‐19
removed from the wafer and embedded on the polydimethylsiloxane. treatment, viral and non‐viral nanoscale vaccine vectors are injected
It has application in nanoinjection of biomolecules into cells and tis- which has undergone minimum 25 years of long development phase
sues [204]. In an investigation, diamond nanoneedles are prepared towards clinical translation. Long term use of nano vaccine is still
with aptamer based nano sensors. This nanoneedle is capable of isolat- not clear due to cost, duration of effectiveness is not understood, incor-
ing NF‐κB from intracellular region [205]. Diamond nanoneedle is cap- poration of new sequence against mutation of virus is not easy and
able of enhancing the delivery of plasmid DNAs into neurons by eight‐ complicated manufacturing and logistics [218].
fold in transfection efficiency compared to lipofection. It is an efficient
method for vector free delivery to cytoplasmic matrix [206]. 7.19. Dry vaccine delivery system

Vaccines are made up of antigens which are usually chemically and


7.16. Ultrasound drug delivery physically unstable in liquid formulation [219,220]. Vaccine delivery
in the dry powder aerosol form is considered as a promising method
Ultrasounds are capable of permeating into the tissue at very low to prevent degradation of macromolecules present in vaccine [221].
amplitude. It can be used to trigger local anesthetic to combat pain In a study, spray freeze dried vaccine was analyzed for anthrax treat-
[207]. Ultrasounds can be used for targeted drug delivery because ment via nasal route. Formulated vaccine can be stored for two and
when a nanoparticle embedded with drug is injected into the blood a half years at room temperature and results suggest it remains as fresh
vessel and irradiated with ultrasonic energy, the nanoparticles expand as intramuscular injection in liquid formulation [222]. In another
and facilitate the release of drug [208]. Ultrasound can be used for study, Norwalk virus‐like particles was incorporated into dry powder
controlled drug delivery. For instance, in a study, tetrodotoxin and of polysaccharide derived from aloe vera for nasal delivery. It was
protoporphyrin IX were encapsulated into micrometric liposomes. observed that nasal mucosal provides large surface area for immune
Exposure of ultrasound showed formation of reactive oxygen species induction. This study concludes that dry vaccine is more immunogenic
(ROS). This ROS reacted with liposome membrane and leads to insta- than liquid formulation [223]. In a recent investigation, nanovaccine
bility and releases tetrodotoxin [209]. (cubosomes with ovalbumin and Quil‐A) was spray dried. Powdered
In a study it was observed that ultrasound can be used to detect gas form vaccine consisted of 10.6 ± 0.7% w/w ovalbumin, among which
vesicles present in the microorganism [210]. The mechanism behind 65% was released with 20 minutes and complete release was recorded
this is that ultrasounds are scattered when comes in contact with gas in 24 hours [224]. Luczo and his colleagues developed powdered form
vesicles which are recorded. But pressure pulses are capable of collaps- of live attenuated influenza vaccines by vapourization and reported
ing the cell which causes disappearance of the ultrasound and can be the minimum loss for 1 year at 25 ℃. And also suggests that this vac-
used for detecting the location of the cell containing gas vesicle [211]. cine can be stored without refrigeration and do not require reconstitu-
In another investigation, ultrasound has also shown to favor the treat- tion and injection during administration [225]. Thus, dried form of
ment of Leptomeningeal metastases in athymic rats. Experiment was vaccine is one of the methods to develop vaccine with improved effi-
divided into three groups: (i) samples with no treatment, (ii) samples ciency and stability.
treated with trastuzumab and (iii) samples were treated with trastuzu-
mab and focused ultrasound combined with micron‐sized stabilized 7.20. Bioceramic nanoparticles
bubbles. Magnetic resonance imaging report showed that group 1, sup-
pressed the tumor cells to 25 ± 17 mm3 and group 3, suppressed it to Bioceramic nanoparticles can be used as a cargo for targeted drug
8 ± 5 mm3 [212]. delivery and prevents enzymatic degradation [226]. Bioceramic
nanoparticles helps in sustained release of drug as reported in a study
7.17. Microchip technology sustained release of gentamicin for 70 hours was recorded poly
(epsilon‐caprolactone) coated calcium phosphate scaffolds [227]. Alu-
Microchip is a micro‐scale device with wireless chipset, sensors and mina, pyrolytic carbons, zirconia, calcium phosphates and silica‐based
drug delivery unit which can be used for pulsatile release of drug glasses or glass ceramics are the commonly known bioceramics [228].
[213]. Electric field can be used to provoke conducting material for Among all the bioceramic NPs, calcium phosphate is widely studied for
controlled release drug [214]. The four ways electric field can provoke drug delivery due to their chemical similarity to biomineralized mam-
drug delivery system are: by opening the pores of conducting material malian tissue leading to a high biocompatibility and inherent
which covers the drug, by shrinking the nanoparticle which ultimately biodegradation [229]. Calcium phosphate possess more reaction sites
release the drug in the presence of electric field, by enhancing the skin for drug binding due to their high surface‐to‐volume ratios compared
permeability and by applying force on ions which enhances the move- to bulk form and high loading capacity [230]. Calcium phosphate NPs
ment of surface‐charged small‐molecule drugs [149]. In a study n‐type can be synthesized via precipitation, sol–gel method, flame‐spray
silicon wafer (resistance of 0.45 ± 0.75 Ω cm and diameter of 100 nm) pyrolysis, solid‐state reactions etc., with different properties, depend-
was used to prepare silicon chip. The efficiency of DNA electroporation ing on morphology, size, and surface properties [231]. In an investiga-
was recorded as 23.08% and 61.21% for neuronal SH‐SY5Y and tion, Platinum (IV) prodrug of cisplatin was loaded into hybrid
epithelial CHO‐K1 cells respectively. It was concluded that silicon nanoparticles of human serum albumin (HSA) and calcium phosphate.

14
A. Sultana et al. Medicine in Drug Discovery 15 (2022) 100134

The pH responsive study showed that, at normal pH the hybrid is to understand the interaction of drug with the environment, involve-
stable and at pH less than or equal to 6, complete release of drug ment in biosystem, biodegradability, disposability and its accumula-
was observed. According to in vitro cytotoxicity analysis, calcium tion [243]. Establishment of regulatory authority is necessary for
phosphate enhanced the cytotoxicity of hybrid [232]. Fig. 8 represent nano‐drug delivery system because along with various benefits of
the schematic illustration of bioceramic nanoparticle used for drug nano‐medicines some studies also suggest the negative impact on
delivery. health. This regulatory body needs to consider ethical, legal and social
impact as well as focus on possible and non‐possible risk of the drug.
7.21. Nanogels Along with environmental impact, ethical impact is also need to be
considered which leads to design of strict rules and regulation. One
Nanogels are 3D nanoparticles obtained by crosslinking hydrophi- of the challenges is communication gap between developed nations
lic or amphiphilic polymer chains which can be used for incorporation and under developed nations is related to risk an assessment and man-
of drug via hydrogen bonding, salt bond formation or hydrophobic agement measure which causes complication in designing standard
interaction [233]. Nanogels reduces toxic effects, enhance therapeutic rules. Another challenge is the knowledge gap among scientist, public,
index of the drug and can be fabricated as stimuli‐responsive materials regulatory body and civil society which lead to inadequate risk evalu-
[234]. It has scope in controlled drug release for cancer therapy such ation and administration [244]. Patenting of nano‐based drug is also
as in an investigation sulfamide‐based zwitterionic monomer was used under confusion whether it should be patented or not. For this reason,
to synthesise nanogels which showed controlled drug release and exhi- Europe has designed a “nanoscience and nanotechnology patent‐
bit prolonged blood circulation [235]. In another study, pH‐responsive monitoring system under European Patent Office [245]. Drug delivery
polypeptide‐based nanogels were developed which was stable at pH system requires proper and systematic rules and regulation followed
7.4 and released doxorubicin under acidic (tumor) condition [236]. by ethical aspects, but currently implementation of such system is
absent due to global governance gap, communication gap and knowl-
edge gap.
7.22. Fullerenes

Fullerenes are allotropes of the carbon and have a structure which


possess sp2 carbons. Fullerenes are useful in achieving controlled 9. Future scope
release of drug to the targeted site, thus have scope for efficient treat-
ment of disease like cancer and HIV [237]. In an investigation por- Drug delivery systems have advanced a lot from the normal basic
phyrin adducts of cyclohexyl‐fullerene were fabricated which pill with unregulated release towards the systems with improved
showed 80% recovery of hypoxia symptoms in one day [238]. Fullere- bioavailability and reduced side effects. Still there are undergoing
nes can also be incorporated with hormone such as erythropoietin and researches focusing on targeted, controlled, monitored and convenient
bioactive component such as warfarin [239,240]. It has antioxidant, drug delivery systems. Researchers need to focus on factors such as
antibacterial and photodynamic effect and also acts as scaffold for drug drug delivery system for future viruses, safety, symbiotic delivery sys-
synthesis [241]. tem, gender sensitive delivery system, affordability, more greener drug
Smart drug delivery system has several advantages among which delivery system and system which resolves unmet clinical needs. These
one of the important factors is, it maintains the drug concentration factors are further discussed in details.
in the organ or tissue but decreases drug dosage frequency. Insufficient
information availability on in vivo trails, cost of drug, problem of
inconsistent pharmacokinetics between preclinical and clinical trials 9.1. Drug delivery for future viruses
are some of the factors which acts as shortcoming of the smart drug
delivery system [242]. Viral infections are one of the health issues which are challenging
to treat due to increased drug resistance and barrier at cellular and
8. Ethics and regulatory affairs in nano-drug delivery system intracellular levels. For example, an effective treatment of herpes sim-
plex virus and herpes zoster are still under study. Frequently encoun-
Variation in characteristics of nano scale particles may cause tered pandemic situation around the world due to Covid‐19 is also an
unwanted effect on environment and human. Therefore it is important example of unavailability of effective drug for treatment. Although
remdesivir was a hope for reducing the recovery of patients but still
many conflicting data is available [246]. Recent development has
led to the focus on nanaotechnology (nanoparticles, liposomes, nano-
spheres, nanogels, nanosuspensions and nanoemulsions) for drug
delivery [247]. For instance, in a study multiwalled carbon nanotubes
conjugated with virus‐specific nanobody and antiviral drug was inves-
tigated for treatment of virus induced central nervous system disease.
It was observed that the mortality of larvae was 100% for control and
27% for treated samples [248]. In an undergoing study, it was
reported that molnupiravir (an antiviral pill) can be used for effective
treatment of gamma, delta and Mu variants of corona virus. Such drugs
are the need for being ready to face future predicted health crisis.

9.2. Safety and efficacy of drug

It is reported that 80% of the drug used for human trail fail to show
Fig. 8. Schematic illustration of bioceramic nanoparticle which acts as a safety and efficacy [249]. Such failure can be overcome by being open
carrier of drug for targeted drug delivery. It consists of target molecule, about the failed trails which also help to speed up the research in drug
surface stabilizing component and drug. development [250].

15
A. Sultana et al. Medicine in Drug Discovery 15 (2022) 100134

9.3. Symbiotic drug delivery ing incorporation of SrCl2 and black phosphorus nanosheets into poly
(lactic‐co‐glycolic acid) [265]. Intelligent drug delivery system is a
Symbiotic drug delivery is an efficient way for targeted and con- potential drug delivery system for future but requires focus on its tox-
trolled drug delivery such as bacteria can be used for treatment of dis- icity, adequate intensity and in vivo evaluation of the performance.
eased human tissue. In a study bacterium was used as vehicle to carry Intelligent drug delivery system will have promising opportunities to
antitumor drug which reduced toxicity and improved delivery effi- attain efficient targeted drug delivery [266].
ciency [251]. In another study, Lactobacillus acidophilus was
microencapsulated in alginate‐gelatin and alginate‐gelatin‐fructooligo 10. Conclusions
saccharides microbeads and results suggested improvement in viability
of microbes in gastrointestinal tract as well as when incorporated in the Drug delivery system plays a vital role in terms of treatment of var-
yogurt [252]. Symbiosis of metal–organic frameworks was fabricated ious health issues. The basic mechanism of drug release involves, bind-
and studied for efficient drug delivery. Drug release profile showed that ing of nanocarrier linked with drug to the receptor, entering the cell
hydrogel release was completed within 5 days whereas, metal–organic via endocytosis and then the drug releases into the endosomes. Tar-
frameworks had sustained release for 7 days [253]. geted delivery of drug can take place either via linker mechanism or
encapsulation. The four basic steps involved in pharmacokinetics once
9.4. Gender sensitive drug delivery the drug is administered are absorption, distribution, metabolism and
elimination of drug.
Gender differences have effect on frequency and severity of phar- This review discusses the various conventional drug delivery sys-
macological side effects [254]. Difference in gender has variation in tems along with their benefits and shortcomings. Drug delivery system
physiological, hormonal, and genetic conditions which may act as a has been evolving very fast with time through implementation of inno-
reason for uneven pharmacological side effects. In a study adverse vative technology. Limitations of the existing or conventional drug
event of antidiabetic drug was investigated based on difference in gen- delivery systems can be overcome with the smart drug delivery system.
der. It was observed that 13 out of 17 system organ class level disor- These smart drug delivery systems (such as nucleic acid‐based, cell‐
ders were common in women than in men [255]. In another study it based, self‐nano emulsifying, self‐micro emulsifying drug delivery sys-
is found that compared to men, women encountered nervous system tems physical and chemical stimuli‐based drug delivery) are capable in
disorders such nausea, headaches, insomnia [256]. Thus, it is neces- improving bioavailability, stability, better absorption and controlled
sary to consider gender differences to design a tailored and more effec- release of drug for effective treatment of health problems. With recent
tive treatment for each individual [257]. advancement novel drug delivery system such as nanoneedle patches,
ultrasound‐based delivery, microchips and many other are widely
9.5. Affordable studied with future perspective for achieving targeted and controlled
drug delivery systems. This study has highlighted the seven major
It has been observed that novel drug with even modest benefits issues which need to be focused for designing of potential and effective
come with the increase price than the previous existing treatment. In drug delivery system and overcoming existing challenges for treatment
a report, four regulatory policies are proposed to induce transparency of health issues.
and reduce unnecessary cost rise of drug. First is, to only allow those
drugs which have added therapeutic benefit. Second is, that all the
Author contributions
drugs should undergo head‐to‐head comparison with other treatments
which include mandatory active‐controlled randomized controlled tri-
A.S collected data for the review article, prepared the initial draft of
als. Third is, that added therapeutic benefit should undergo indirect
the manuscript, and worked with the co‐authors to edit and refine the
comparisons. And the last step is to describe the comparison of a coun-
manuscript before submission. M.Z., V.T., and T.S.S.K. revised and edi-
terfactual scenario with the bad or good effects of treatment [258].
ted the manuscript. S.R. and M.Z. supervised the work, revised the
manuscript, and approved the final version to be published. All
9.6. Greener drug delivery system
authors have read and agreed to the published version of the
manuscript.
Drug delivery fabricated using living organisms, biomolecules and
environmentally friendly are greener compared to the existing drug
delivery systems. For instance, oligonucleotide (polynucleotide DNA Funding
or RNA chain) acts as a aptamer and could be used for treatment
due to its low adverse effect, high specificity, least chance for resis- The authors received NO specific funding for this work.
tance development and minimum cytotoxicity [259]. Natural compo-
nents such as curcumin, camptothecin, anthracyclins and collagen Ethical statement
can be incorporated into nanofiber via electrospinning and used for
treatment of cancer [260,261]. This study was conducted with integrity, fidelity, and honesty. No
humans or animals were harmed.
9.7. Intelligent drug delivery system
Conflicts of interest
Intelligent drug delivery system involves the delivery of drug by
sensing the change based on stimuli [262].Intelligent drug delivery The authors declare that there are no conflicts of interest.
helps in achieving targeted drug delivery via pH responsive, light sen-
sitivity, redox responsive, enzyme responsive, thermo responsive and
References
other responsive systems such as saccharide sensitive system, urea
responsive system, electro responsive system and may other [263]. [1] Tibbitt MW, Dahlman JE, Langer R. Emerging frontiers in drug delivery. J Am
For instance in a study mesoporous silica nanoparticles were coated Chem Soc, 138, 704–17.
with poly(ethylene glycol) and poly(2‐(pentamethyleneimino)ethyl [2] Li C, Wang J, Wang Y, Gao H, Wei G, Huang Y, Yu H, Gan Y, Wang Y, Mei L, Chen
H. Recent progress in drug delivery. Acta Pharm Sin B., 9, 1145-62.
methacrylate) to obtain pH triggered targeted drug delivery [264]. [3] Holowka E, Bhatia SK. Drug delivery. Springer-Verlag New York; 2016.
Near infrared light targeted drug delivery is reported in a study involv- doi:10.1007/978-1-4939-1998-7.

16
A. Sultana et al. Medicine in Drug Discovery 15 (2022) 100134

[4] Zhang L, Zhang L, Zhang M, Pang Y, Li Z, Zhao A, et al. Self-emulsifying drug [38] Golan DE, Tashjian AH, Armstrong EJ, editors. Principles of pharmacology: the
delivery system and the applications in herbal drugs. Drug Delivery pathophysiologic basis of drug therapy. Lippincott Williams & Wilkins; 2011 Dec
2015;22:475–86. 15.
[5] Sinha VR, Bansal K, Kaushik R, Kumria R, Trehan A. Poly-ϵ-caprolactone [39] Valikhani D, Bolivar JM, Pelletier JN. An overview of cytochrome P450
microspheres and nanospheres: an overview. Int J Pharm 2004;278:1–23. immobilization strategies for drug metabolism studies, biosensing, and
[6] Sahoo D, Bandaru R, Samal SK, Naik R, Kumar P, Kesharwani P, Dandela R. Oral biocatalytic applications: challenges and opportunities. ACS Catal
drug delivery of nanomedicine. In Theory and Applications of Nonparenteral 2021;11:9418–34.
Nanomedicines, 2021, 1 (pp. 181-207). Academic Press. [40] Harada N, Miwa GT, Walsh JS, Lu AY. Kinetic isotope effects on cytochrome P-
[7] Saltzman WM, Torchilin VP. Drug delivery systems. AccessScience, McGraw-Hill 450-catalyzed oxidation reactions. Evidence for the irreversible formation of an
Companies; 2008. activated oxygen intermediate of cytochrome P-448. J Biol Chem
[8] Wong PT, Choi SK. Mechanisms of drug release in nanotherapeutic delivery 1984;259:3005–10.
systems. Chem Rev 2015;115:3388–432. [41] Zhang Z, Tang W. Drug metabolism in drug discovery and development. Acta
[9] Caparco AA, Bommarius AS, Champion JA. Effect of peptide linker length and Pharm Sin B 2018;8:721–32.
composition on immobilization and catalysis of leucine zipper-enzyme fusion [42] Meibohm B, Evans WE. Clinical pharmacodynamics and pharmacokinetics.
proteins. AIChE J 2018;64:2934–46. Textbook of Therapeutics: Drug and Disease Management. 8th ed. Philadelphia:
[10] Reddy Chichili VP, Kumar V, Sivaraman J. Linkers in the structural biology of Lippincott Williams & Wilkins, 2006, 1-30.
protein–protein interactions. Protein Sci 2013;22:153–67. [43] Currie GM. Pharmacology, part 2: introduction to pharmacokinetics. J Nucl Med
[11] Bargh JD, Isidro-Llobet A, Parker JS, Spring DR. Cleavable linkers in Technol 2018;46:221–30.
antibody–drug conjugates. Chem Soc Rev 2019;48:4361–74. [44] Choi YH, Han HK. Nanomedicines: current status and future perspectives in
[12] Llopis J, McCaffery JM, Miyawaki A, Farquhar MG, Tsien RY. Measurement of aspect of drug delivery and pharmacokinetics. J Pharm Invest 2018;48(1):43–60.
cytosolic, mitochondrial, and Golgi pH in single living cells with green [45] Salahudeen MS, Nishtala PS. An overview of pharmacodynamic modelling,
fluorescent proteins. Proc Natl Acad Sci 1998;95:6803–8. ligand-binding approach and its application in clinical practice. Saudi Pharm J
[13] Testa B, Mayer JM, Mayer J. Hydrolysis in drug and prodrug metabolism. John 2017;25:165–75.
Wiley & Sons; 2003. [46] Liu E, Zhang M, Huang Y. Pharmacokinetics and pharmacodynamics (PK/PD) of
[14] Yang J, Chen H, Vlahov IR, Cheng JX, Low PS. Evaluation of disulfide reduction bionanomaterials. Biomed Nanomater 2016;18:1–60.
during receptor-mediated endocytosis by using FRET imaging. Proc Natl Acad Sci [47] Goutelle S, Maurin M, Rougier F, Barbaut X, Bourguignon L, Ducher M, et al. The
2006;103:13872–7. Hill equation: a review of its capabilities in pharmacological modelling. Fundam
[15] Paulsen CE, Carroll KS. Cysteine-mediated redox signaling: chemistry, biology, Clin Pharmacol 2008;22:633–48.
and tools for discovery. Chem Rev 2013;113:4633–79. [48] Sastry SV, Nyshadham JR, Fix JA. Recent technological advances in oral drug
[16] Lee MH, Yang Z, Lim CW, Lee YH, Dongbang S, Kang C, et al. Disulfide-cleavage- delivery–a review. Pharm Sci Technol Today 2000;3:138–45.
triggered chemosensors and their biological applications. Chem Rev [49] Patel MR, Patel RB, Thakore SD. Nanoemulsion in drug delivery. In Applications of
2013;113:5071–109. nanocomposite materials in drug delivery 2018, 667–700, Woodhead Publishing.
[17] Kuśmierek K, Chwatko G, Głowacki R, Bald E. Determination of endogenous [50] Alexander A, Tripathi DK, Verma T, Maurya J, Patel S. Mechanism responsible for
thiols and thiol drugs in urine by HPLC with ultraviolet detection. J Chromatogr B mucoadhesion of mucoadhesive drug delivery system: a review. 2011.
2009;877:3300–8. [51] Ns M, Ojha A, Tyagi Y, Negi M. Mucoadhesion: a novelistic platform for drug
[18] Bailey KM, Wojtkowiak JW, Hashim AI, Gillies RJ. In Advances in Biochemical delivery system. Int J Pharm Drug Anal 2014:773–81.
Pharmacology; Keiran, S. M. S., Ed.;Academic Press: Waltham, MA, 2012, 65. [52] Patel AR, Patel DA, Chaudhry SV. Mucoadhesive buccal drug delivery system. Int
[19] Wilson WR, Hay MP. Targeting hypoxia in cancer therapy. Nat Rev Cancer J Pharm Life Sci 2011;2.
2011;11:393–410. [53] Xu J, Tam M, Samaei S, Lerouge S, Barralet J, Stevenson MM, et al. Mucoadhesive
[20] Wong AD, DeWit MA, Gillies ER. Amplified release through the stimulus triggered chitosan hydrogels as rectal drug delivery vessels to treat ulcerative colitis. Acta
degradation of self-immolative oligomers, dendrimers, and linear polymers. Adv Biomater 2017;48:247–57.
Drug Deliv Rev 2012;64:1031–45. [54] Purohit TJ, Hanning SM, Wu Z. Advances in rectal drug delivery systems. Pharm
[21] Houba PH, Boven E, Van der Meulen-Muileman IH, Leenders RG, Scheeren JW, Dev Technol 2018;23:942–52.
Pinedo HM, et al. A novel doxorubicin-glucuronide prodrug DOX-GA3 for [55] Surti N, Mahajan A, Misra A. Polymers in Rectal Drug Delivery. In Applications of
tumour-selective chemotherapy: distribution and efficacy in experimental human Polymers in Drug Delivery, 2021, 263-280, Elsevier.
ovarian cancer. Br J Cancer 2001;84:550–7. [56] Daniel A. Haas, 38 - Management of Fear and Anxiety, Editor(s): Frank J. Dowd,
[22] Klotz U. The pharmacological profile and clinical use of mesalazine (5- Barton S. Johnson, Angelo J. Mariotti, Pharmacology and Therapeutics for Dentistry
aminosalicylic acid). Arzneimittelforschung 2012;62:53–8. (Seventh Edition), Mosby, 2017, 575-583, ISBN 9780323393072, DOI:
[23] Lakatos PL, Lakatos L. Once daily 5-aminosalicylic acid for the treatment of https://doi.org/10.1016/B978-0-323-39307-2.00038-2.
ulcerative colitis; are we there yet? Pharmacol Res 2008;58:90–5. [57] Jain KK. An overview of drug delivery systems. Drug Deliv Syst 2020:1–54.
[24] Zeng F, Zimmerman SC. Dendrimers in supramolecular chemistry: from [58] Kim H, Park H, Lee SJ. Effective method for drug injection into subcutaneous
molecular recognition to self-assembly. Chem Rev 1997;97:1681–712. tissue. Sci Rep 2017;7. 1–1.
[25] Brunsveld L, Folmer BJ, Meijer EW, Sijbesma RP. Supramolecular polymers. [59] Gutierrez JJ, Intramuscular MS. Injection. StatPearls [Internet] 2020.
Chem Rev 2001;101:4071–98. [60] Grassin-Delyle S, Buenestado A, Naline E, Faisy C, Blouquit-Laye S, Couderc LJ,
[26] Mura S, Nicolas J, Couvreur P. Stimuli-responsive nanocarriers for drug delivery. et al. Intranasal drug delivery: an efficient and non-invasive route for systemic
Nat Mater 2013;12:991–1003. administration: focus on opioids. Pharmacol Ther 2012;134:366–79.
[27] Gao Y, Chen Y, Ji X, He X, Yin Q, Zhang Z, et al. Controlled intracellular release of [61] Paranjpe M, Müller-Goymann CC. Nanoparticle-mediated pulmonary drug
doxorubicin in multidrug-resistant cancer cells by tuning the shell-pore sizes of delivery: a review. Int J Mol Sci 2014;15:5852–73.
mesoporous silica nanoparticles. ACS Nano 2011;5:9788–98. [62] Sonia TA, Sharma CP, Routes of administration of insulin, In Woodhead
[28] Kenakin T. A pharmacology primer: techniques for more effective and strategic Publishing Series in Biomedicine, Oral Delivery of Insulin, Woodhead Publishing,
drug discovery. Academic Press 2018;26:245–93. 2014, Pages 59-112, ISBN 9781907568473, DOI: https://doi.org/10.1533/
[29] Buckley ST, Fischer SM, Fricker G, Brandl M. In vitro models to evaluate the 9781908818683.59.
permeability of poorly soluble drug entities: challenges and perspectives. Eur J [63] Kumar A, Montemagno C, Choi HJ. Smart microparticles with a pH-responsive
Pharm Sci 2012;45:235–50. macropore for targeted oral drug delivery. Sci Rep 2017;7:1–5.
[30] Devadasu VR, Deb PK, Maheshwari R, Sharma P, Tekade RK. Physicochemical, [64] Hu S, Pei X, Duan L, Zhu Z, Liu Y, Chen J, et al. A mussel-inspired film for
pharmaceutical, and biological considerations in GIT absorption of drugs. In adhesion to wet buccal tissue and efficient buccal drug delivery. Nat Commun
Dosage Form Design Considerations, 2018, 149–178. Academic Press. 2021;12:1–7.
[31] Su C, Liu Y, Li R, Wu W, Fawcett JP, Gu J. Absorption, distribution, metabolism [65] Le Bras A. Local drug delivery to brain tumor. Lab Anim 2020;49:18. https://doi.
and excretion of the biomaterials used in Nanocarrier drug delivery systems. Adv org/10.1038/s41684-019-0453-0.
Drug Deliv Rev 2019;143:97–114. [66] Lapin NA, Gill K, Shah BR, Chopra R. Consistent opening of the blood brain
[32] Bok M, Zhao ZJ, Jeon S, Jeong JH, Lim E. Ultrasonically and iontophoretically barrier using focused ultrasound with constant intravenous infusion of
enhanced drug-delivery system based on dissolving microneedle patches. Sci Rep microbubble agent. Sci Rep 2020;10. 1–1.
2020;10:1. [67] Badkar AV, Gandhi RB, Davis SP, LaBarre MJ. Subcutaneous delivery of high-
[33] Viswanathan P, Muralidaran Y, Ragavan G. Challenges in oral drug delivery: a dose/volume biologics: current status and prospect for future advancements.
nano-based strategy to overcome. InNanostructures for oral medicine, 2017, 173- Drug Des Dev Ther 2021;15:159.
201. Elsevier. [68] Zhang Y, Fan D, Zhang Y, Zhang S, Wang H, Liu Z, et al. Using corneal confocal
[34] Malik S, Subramanian S, Hussain T, Nazir A, Ramakrishna S. Electrosprayed microscopy to compare Mecobalamin intramuscular injections vs oral tablets in
nanoparticles as drug delivery systems for biomedical applications. Curr Pharm treating diabetic peripheral neuropathy: a RCT. Sci Rep 2021;11:1.
Des 2021. [69] Kim D, Kim YH, Kwon S. Enhanced nasal drug delivery efficiency by increasing
[35] Gao X, Gu X, Chen H. The Distribution and Elimination of Nanomaterials in Brain. mechanical loading using hypergravity. Sci Rep 2018;8:1–8.
InNeurotoxicity of nanomaterials and nanomedicin, 2017, 59-74. Academic Press. [70] Mohseni M, Gilani K, Mortazavi SA. Preparation and characterization of rifampin
[36] Lammers T, Koczera P, Fokong S, Gremse F, Ehling J, Vogt M, et al. Theranostic loaded mesoporous silica nanoparticles as a potential system for pulmonary drug
USPIO-loaded microbubbles for mediating and monitoring blood-brain barrier delivery. Iran J Pharm Res: IJPR 2015;14:27.
permeation. Adv Funct Mater 2015;25:36–43. [71] Lahiji SF, Dangol M, Jung H. A patchless dissolving microneedle delivery system
[37] Waller D, Renwick A, Hillier K. Medical pharmacology and therapeutics. 2nd enabling rapid and efficient transdermal drug delivery. Sci Rep 2015;5:1–7.
ed. London: Elsevier; 2006.

17
A. Sultana et al. Medicine in Drug Discovery 15 (2022) 100134

[72] Alvandi N, Rajabnejad M, Taghvaei Z, Esfandiari N. New generation of viral [102] Das MP, Pandey G, Neppolian B, Das J. Design of poly-l-glutamic acid embedded
nanoparticles for targeted drug delivery in cancer therapy. J Drug Target mesoporous bioactive glass nanospheres for pH-stimulated chemotherapeutic
2021:1–5. drug delivery and antibacterial susceptibility. Colloids Surf, B 2021;202:111700.
[73] Müller RH, Mäder K, Gohla S. Solid lipid nanoparticles (SLN) for controlled drug [103] Samrot AV, Burman U, Philip SA, Shobana N, Chandrasekaran K. Synthesis of
delivery–a review of the state of the art. Eur J Pharm Biopharm 2000;50:161–77. curcumin loaded polymeric nanoparticles from crab shell derived chitosan for
[74] Malam Y, Loizidou M, Seifalian AM. Liposomes and nanoparticles: nanosized drug delivery. Inf Med Unlocked 2018;10:159–82.
vehicles for drug delivery in cancer. Trends Pharmacol Sci 2009;30:592–9. [104] Palanikumar L, Al-Hosani S, Kalmouni M, Nguyen VP, Ali L, Pasricha R, et al. pH-
[75] Duan Y, Dhar A, Patel C, Khimani M, Neogi S, Sharma P, et al. A brief review on responsive high stability polymeric nanoparticles for targeted delivery of
solid lipid nanoparticles: Part and parcel of contemporary drug delivery systems. anticancer therapeutics. Commun Biol 2020;3:1–7.
RSC Adv 2020;10:26777–91. [105] Joseph A, Wood T, Chen CC, Corry K, Snyder JM, Juul SE, et al. Curcumin-loaded
[76] Smith T, Affram K, Nottingham EL, Han B, Amissah F, Krishnan S, et al. polymeric nanoparticles for neuroprotection in neonatal rats with hypoxic-
Application of smart solid lipid nanoparticles to enhance the efficacy of 5- ischemic encephalopathy. Nano Res 2018;11:5670–88.
fluorouracil in the treatment of colorectal cancer. Sci Rep 2020;10:1–4. https:// [106] Salama AH, AbouSamra MM, Awad GE, Mansy SS. Promising bioadhesive
doi.org/10.1038/s41598-020-73218-6. ofloxacin-loaded polymeric nanoparticles for the treatment of ocular
[77] Parvez S, Yadagiri G, Gedda MR, Singh A, Singh OP, Verma A, et al. Modified inflammation: Formulation and in vivo evaluation. Drug Deliv Transl Res
solid lipid nanoparticles encapsulated with Amphotericin B and Paromomycin: an 2021;11:1943–57.
effective oral combination against experimental murine visceral leishmaniasis. [107] Zhu Y, Liao L. Applications of nanoparticles for anticancer drug delivery: a
Sci Rep 2020;10:1–4. review. J Nanosci Nanotechnol 2015;15:4753–73.
[78] Campos EV, de Oliveira JL, da Silva CM, Pascoli M, Pasquoto T, Lima R, et al. [108] Tsuji A, Nguyen TL, Mizoue Y, Ishihara K, Yusa SI. Water-soluble polymer
Polymeric and solid lipid nanoparticles for sustained release of carbendazim and micelles formed from amphiphilic diblock copolymers bearing pendant
tebuconazole in agricultural applications. Sci Rep 2015;5:1–4. phosphorylcholine and methoxyethyl groups. Polym J 2021;53:805–14.
[79] Banerjee I, De M, Dey G, Bharti R, Chattopadhyay S, Ali N, et al. A peptide- [109] Sanati S, Taghavi S, Abnous K, Taghdisi SM, Babaei M, Ramezani M, et al.
modified solid lipid nanoparticle formulation of paclitaxel modulates immunity Fabrication of anionic dextran-coated micelles for aptamer targeted delivery of
and outperforms dacarbazine in a murine melanoma model. Biomater Sci camptothecin and survivin-shRNA to colon adenocarcinoma. Gene Ther
2019;7:1161–78. 2021:1–4.
[80] Xie S, Yang F, Tao Y, Chen D, Qu W, Huang L, et al. Enhanced intracellular [110] Hu J, Miura S, Na K, Bae YH. pH-responsive and charge shielded cationic micelle
delivery and antibacterial efficacy of enrofloxacin-loaded docosanoic acid solid of poly (L-histidine)-block-short branched PEI for acidic cancer treatment. J
lipid nanoparticles against intracellular Salmonella. Sci Rep 2017;7:1–9. Control Release 2013;172:69–76.
[81] Parhi P, Suklabaidya S, Sahoo SK. Enhanced anti-metastatic and anti-tumorigenic [111] Deepagan VG, Kwon S, You DG, Um W, Ko H, Lee H, et al. In situ diselenide-
efficacy of Berbamine loaded lipid nanoparticles in vivo. Sci Rep 2017;7:1–3. crosslinked polymeric micelles for ROS-mediated anticancer drug delivery.
[82] Ban C, Park JB, Cho S, Kim HR, Kim YJ, Choi YJ, et al. Reduction of focal Biomaterials 2016;103:56–66.
sweating by lipid nanoparticle-delivered myricetin. Sci Rep 2020;10:1–4. [112] Teo JY, Chin W, Ke X, Gao S, Liu S, Cheng W, et al. pH and redox dual-responsive
[83] ud Din F, Aman W, Ullah I, Qureshi OS, Mustapha O, Shafique S, Zeb A. Effective biodegradable polymeric micelles with high drug loading for effective anticancer
use of nanocarriers as drug delivery systems for the treatment of selected tumors. drug delivery. Nanomed Nanotechnol Biol Med 2017;13:431–42.
Int J Nanomed, 2017, 12:7291. [113] Chen WH, Luo GF, Lei Q, Jia HZ, Hong S, Wang QR, et al. MMP-2 responsive
[84] Dimov N, Kastner E, Hussain M, Perrie Y, Szita N. Formation and purification of polymeric micelles for cancer-targeted intracellular drug delivery. Chem
tailored liposomes for drug delivery using a module-based micro continuous-flow Commun 2015;51:465–8.
system. Sci Rep 2017;7:1–3. [114] He B, Hu HY, Tan T, Wang H, Sun KX, Li YP, et al. IR-780-loaded polymeric
[85] Deng W, Chen W, Clement S, Guller A, Zhao Z, Engel A, et al. Controlled gene and micelles enhance the efficacy of photothermal therapy in treating breast cancer
drug release from a liposomal delivery platform triggered by X-ray radiation. Nat lymphatic metastasis in mice. Acta Pharmacol Sin 2018;39:132–9.
Commun 2018;9. 1–1. [115] Manchester M, Singh P. Virus-based nanoparticles (VNPs): platform technologies
[86] Hynynen K. Hyperthermia-induced drug delivery in humans. Nat Biomed Eng for diagnostic imaging. Adv Drug Deliv Rev 2006;58:1505–22.
2018;2:637–9. [116] Singh P, Prasuhn D, Yeh RM, Destito G, Rae CS, Osborn K, et al. Bio-distribution,
[87] Miao YQ, Chen MS, Zhou X, Guo LM, Zhu JJ, Wang R, et al. Chitosan toxicity and pathology of cowpea mosaic virus nanoparticles in vivo. J Control
oligosaccharide modified liposomes enhance lung cancer delivery of paclitaxel. Release 2007;120.
Acta Pharmacol Sin 2021;19:1–9. [117] Chauhan N, Wang X. Nanocages for virus inhibition. Nat Mater 2021;20:1176–7.
[88] Yoo H, Park JS, Oh SS, Kang H. Osmotically balanced, large unilamellar [118] Ghosh S, Banerjee M. A smart viral vector for targeted delivery of hydrophobic
liposomes that enable sustained bupivacaine release for prolonged pain relief in drugs. Sci Rep 2021;11. 1–1.
in vivo rat models. Sci Rep 2021;11:1–9. [119] Chariou PL, Lee KL, Wen AM, Gulati NM, Stewart PL, Steinmetz NF. Detection
[89] Wang Z, Zhao Y, Jiang Y, Lv W, Wu L, Wang B, et al. Enhanced anti-ischemic and imaging of aggressive cancer cells using an epidermal growth factor receptor
stroke of ZL006 by T7-conjugated PEGylated liposomes drug delivery system. Sci (EGFR)-targeted filamentous plant virus-based nanoparticle. Bioconjug Chem
Rep 2015;5:1–5. 2015;26:262–9.
[90] Zhao Z, Zhao Y, Xie C, Chen C, Lin D, Wang S, et al. Dual-active targeting [120] Biabanikhankahdani R, Alitheen NB, Ho KL, Tan WS. pH-responsive virus-like
liposomes drug delivery system for bone metastatic breast cancer: Synthesis and nanoparticles with enhanced tumour-targeting ligands for cancer drug delivery.
biological evaluation. Chem Phys Lipids 2019;223:104785. Sci Rep 2016;6:1–3.
[91] Du Y, He W, Xia Q, Zhou W, Yao C, Li X. Thioether phosphatidylcholine [121] Pitek AS, Wang Y, Gulati S, Gao H, Stewart PL, Simon DI, et al. Elongated plant
liposomes: a novel ROS-responsive platform for drug delivery. ACS Appl Mater virus-based nanoparticles for enhanced delivery of thrombolytic therapies. Mol
Interfaces 2019;11:37411–20. Pharm 2017;14:3815–23.
[92] Lyu Z, Peng L. Potent drugless dendrimers. Nat Biomed Eng 2017;1:686–8. [122] Finbloom JA, Aanei IL, Bernard JM, Klass SH, Elledge SK, Han K, et al. Evaluation
[93] Somani S, Laskar P, Altwaijry N, Kewcharoenvong P, Irving C, Robb G, et al. of three morphologically distinct virus-like particles as nanocarriers for
PEGylation of polypropylenimine dendrimers: effects on cytotoxicity, DNA convection-enhanced drug delivery to glioblastoma. Nanomaterials
condensation, gene delivery and expression in cancer cells. Sci Rep 2018;8:1–3. 2018;8:1007. https://doi.org/10.3390/nano8121007.
[94] Moradian C, Rahbarizadeh F. PE38-based gene therapy of HER2-positive breast [123] Shukla S, Roe AJ, Liu R, Veliz FA, Commandeur U, Wald DN, et al. Affinity of
cancer stem cells via VHH-redirected polyamidoamine dendrimers. Sci Rep plant viral nanoparticle potato virus X (PVX) towards malignant B cells enables
2021;11:1–5. cancer drug delivery. Biomater Sci 2020;8:3935–43.
[95] Benseny-Cases N, Álvarez-Marimon E, Aso E, Carmona M, Klementieva O, [124] Hu H, Steinmetz NF. Cisplatin prodrug-loaded nanoparticles based on physalis
Appelhans D, et al. In situ identification and G4-PPI-His-Mal-dendrimer-induced mottle virus for cancer therapy. Mol Pharm 2020;17:4629–36.
reduction of early-stage amyloid aggregates in Alzheimer’s disease transgenic [125] Bianco A. Carbon nanotubes for the delivery of therapeutic molecules. Expert
mice using synchrotron-based infrared imaging. Sci Rep 2021;11:1–9. Opin Drug Deliv 2004;1:57–65.
[96] Wu SY, Chou HY, Yuh CH, Mekuria SL, Kao YC, Tsai HC. Radiation-sensitive [126] Madani SY, Naderi N, Dissanayake O, Tan A, Seifalian AM. A new era of cancer
dendrimer-based drug delivery system. Adv Sci 2018;5:1700339. treatment: carbon nanotubes as drug delivery tools. Int J Nanomed 2011;6:2963.
[97] England RM, Moss JI, Gunnarsson A, Parker JS, Ashford MB. Synthesis and [127] Kunisaki A, Kodama A, Ishikawa M, Ueda T, Lima MD, Kondo T, et al. Carbon-
characterization of dendrimer-based polysarcosine star polymers: well-defined, nanotube yarns induce axonal regeneration in peripheral nerve defect. Sci Rep
versatile platforms designed for drug-delivery applications. Biomacromolecules 2021;11. 1-0.
2020;21:3332–41. [128] Maleki R, Afrouzi HH, Hosseini M, Toghraie D, Piranfar A, Rostami S. pH-
[98] Chittasupho C, Anuchapreeda S, Sarisuta N. CXCR4 targeted dendrimer for anti- sensitive loading/releasing of doxorubicin using single-walled carbon nanotube
cancer drug delivery and breast cancer cell migration inhibition. Eur J Pharm and multi-walled carbon nanotube: A molecular dynamics study. Comput
Biopharm 2017;119:310–21. Methods Programs Biomed 2020;186:105210.
[99] Prabhu RH, Patravale VB, Joshi MD. Polymeric nanoparticles for targeted [129] Ozden S, Macwan IG, Owuor PS, Kosolwattana S, Autreto PA, Silwal S, et al.
treatment in oncology: current insights. Int J Nanomed 2015;10:1001. Bacteria as bio-template for 3D carbon nanotube architectures. Sci Rep
[100] Zhao J, Yang H, Li J, Wang Y, Wang X. Fabrication of pH-responsive PLGA 2017;7:1–8.
(UCNPs/DOX) nanocapsules with upconversion luminescence for drug delivery. [130] Dehaghani MZ, Yousefi F, Seidi F, Bagheri B, Mashhadzadeh AH, Naderi G, et al.
Sci Rep 2017;7. 1–1. Encapsulation of an anticancer drug Isatin inside a host nano-vehicle SWCNT: a
[101] Cadete A, Olivera A, Besev M, Dhal PK, Gonçalves L, Almeida AJ, et al. Self- molecular dynamics simulation. Sci Rep 2021;11:1–10.
assembled hyaluronan nanocapsules for the intracellular delivery of anticancer
drugs. Sci Rep 2019;9. 1–1.

18
A. Sultana et al. Medicine in Drug Discovery 15 (2022) 100134

[131] Maleki R, Afrouzi HH, Hosseini M, Toghraie D, Rostami S. Molecular dynamics [165] Cutler JI, Zhang K, Zheng D, Auyeung E, Prigodich AE, Mirkin CA. Polyvalent
simulation of Doxorubicin loading with N-isopropyl acrylamide carbon nanotube nucleic acid nanostructures. J Am Chem Soc 2011;133:9254–7.
in a drug delivery system. Comput Methods Programs Biomed 2020;184:105303. [166] Keener AB. How extracellular vesicles can enhance drug delivery. Nature
[132] Gangrade A, Mandal BB. Injectable carbon nanotube impregnated silk based 2020;582:7812.
multifunctional hydrogel for localized targeted and on-demand anticancer drug [167] Dai J, Su Y, Zhong S, Cong L, Liu B, Yang J, et al. Exosomes: Key players in cancer
delivery. ACS Biomater Sci Eng 2019;5:2365–81. and potential therapeutic strategy. Signal Transd Target Ther 2020;5:1.
[133] Tan JM, Bullo S, Fakurazi S, Hussein MZ. Preparation, characterisation and [168] Zhang W, Wang Q, Yang Y, Zhou S, Zhang P, Feng T. The role of exosomal
biological evaluation of biopolymer-coated multi-walled carbon nanotubes for lncRNAs in cancer biology and clinical management. Exp Mol Med
sustained-delivery of silibinin. Sci Rep 2020;10:1–5. 2021;53:1669–73.
[134] Andhari SS, Wavhale RD, Dhobale KD, Tawade BV, Chate GP, Patil YN, et al. Self- [169] Liang Y, Duan L, Lu J, Xia J. Engineering exosomes for targeted drug delivery.
propelling targeted magneto-nanobots for deep tumor penetration and pH- Theranostics 2021;11:3183.
responsive intracellular drug delivery. Sci Rep 2020;10:1–6. [170] Witzigmann D, Kulkarni JA, Leung J, Chen S, Cullis PR, van der Meel R. Lipid
[135] Yoosefian M, Jahani M. A molecular study on drug delivery system based on nanoparticle technology for therapeutic gene regulation in the liver. Adv Drug
carbon nanotube for the novel norepinephrine prodrug, Droxidopa. J Mol Liq Deliv Rev 2020;159:344–63.
2019;284:258–64. [171] García-Pinel B, Porras-Alcalá C, Ortega-Rodríguez A, Sarabia F, Prados J,
[136] Saeednia L, Yao L, Cluff K, Asmatulu R. Sustained releasing of methotrexate from Melguizo C, et al. Lipid-based nanoparticles: application and recent advances
injectable and thermosensitive chitosan–carbon nanotube hybrid hydrogels in cancer treatment. Nanomaterials 2019;9:638.
effectively controls tumor cell growth. ACS Omega 2019;4:4040–8. [172] Ganesan P, Narayanasamy D. Lipid nanoparticles: Different preparation
[137] Liu X, Xu D, Liao C, Fang Y, Guo B. Development of a promising drug delivery for techniques, characterization, hurdles, and strategies for the production of solid
formononetin: Cyclodextrin-modified single-walled carbon nanotubes. J Drug lipid nanoparticles and nanostructured lipid carriers for oral drug delivery.
Delivery Sci Technol 2018;43:461–8. Sustainable Chem Pharm 2017;6:37–56.
[138] Slowing II, Vivero-Escoto JL, Wu CW, Lin VS. Mesoporous silica nanoparticles as [173] Sargent J. Aptamer–lipid nanoparticle conjugates for RNAi in bone. Nat Rev
controlled release drug delivery and gene transfection carriers. Adv Drug Deliv Endocrinol 2015;11:194.
Rev 2008;60:1278–88. [174] He F, Wen N, Xiao D, Yan J, Xiong H, Cai S, et al. Aptamer-based targeted drug
[139] Watermann A, Brieger J. Mesoporous silica nanoparticles as drug delivery delivery systems: Current potential and challenges. Curr Med Chem
vehicles in cancer. Nanomaterials 2017;7:189. 2020;27:2189–219.
[140] Manzano M, Vallet-Regí M. Mesoporous silica nanoparticles for drug delivery. [175] Blind M, Blank M. Aptamer selection technology and recent advances. Mol Ther-
Adv Funct Mater 2020;30:1902634. Nucleic Acids 2015;4:e223.
[141] Vallet-Regí M, Colilla M, Izquierdo-Barba I, Manzano M. Mesoporous silica [176] Huang YF, Shangguan D, Liu H, Phillips JA, Zhang X, Chen Y, et al. Molecular
nanoparticles for drug delivery: Current insights. Molecules 2018;23:47. assembly of an aptamer-drug conjugate for targeted drug delivery to tumor cells.
[142] Chen M, Hu J, Wang L, Li Y, Zhu C, Chen C, et al. Targeted and redox-responsive Chembiochem 2009;10:862.
drug delivery systems based on carbonic anhydrase IX-decorated mesoporous [177] Alshaer W, Hillaireau H, Fattal E. Aptamer-guided nanomedicines for anticancer
silica nanoparticles for cancer therapy. Sci Rep 2020;10:1–2. drug delivery. Adv Drug Deliv Rev 2018;134:122–37.
[143] Zhang BB, Chen XJ, Fan XD, Zhu JJ, Wei YH, Zheng HS, et al. Lipid/PAA-coated [178] Ouyang C, Zhang S, Xue C, Yu X, Xu H, Wang Z, et al. Precision-guided missile-
mesoporous silica nanoparticles for dual-pH-responsive codelivery of arsenic like DNA nanostructure containing warhead and guidance control for aptamer-
trioxide/paclitaxel against breast cancer cells. Acta Pharmacol Sin based targeted drug delivery into cancer cells in vitro and in vivo. J Am Chem Soc
2021;42:832–42. 2020;142:1265–77.
[144] LaBauve AE, Rinker TE, Noureddine A, Serda RE, Howe JY, Sherman MB, et al. [179] Li T, Dong H, Zhang C, Mo R. Cell-based drug delivery systems for biomedical
Lipid-coated mesoporous silica nanoparticles for the delivery of the ML336 applications. Nano Res 2018;11:5240–57.
antiviral to inhibit encephalitic alphavirus infection. Sci Rep 2018;8:1–3. [180] Millan CG, Marinero ML, Castaneda AZ, Lanao JM. Drug, enzyme and peptide
[145] Naz S, Wang M, Han Y, Hu B, Teng L, Zhou J, et al. Enzyme-responsive delivery using erythrocytes as carriers. J Control Release 2004;95:27–49.
mesoporous silica nanoparticles for tumor cells and mitochondria multistage- [181] Zakrzewski W, Dobrzyński M, Szymonowicz M, Rybak Z. Stem cells: past,
targeted drug delivery. Int J Nanomed 2019;14:2533. present, and future. Stem Cell Res Ther 2019;10:1–22.
[146] Shah A, Aftab S, Nisar J, Ashiq MN, Iftikhar FJ. Nanocarriers for targeted drug [182] Aboody KS, Brown A, Rainov NG, Bower KA, Liu S, Yang W, et al. Neural stem
delivery. J Drug Delivery Sci Technol 2021;62:102426. cells display extensive tropism for pathology in adult brain: evidence from
[147] Huda S, Alam MA, Sharma PK. Smart nanocarriers-based drug delivery for cancer intracranial gliomas. Proc Natl Acad Sci 2000;97:12846–51.
therapy: An innovative and developing strategy. J Drug Delivery Sci Technol [183] Evangelopoulos M, Yazdi IK, Acciardo S, Palomba R, Giordano F, Pasto A, et al.
2020;60:102018. Biomimetic cellular vectors for enhancing drug delivery to the lungs. Sci Rep
[148] Saleem K, Khursheed Z, Hano C, Anjum I, Anjum S. Applications of nanomaterials 2020;10. 1-0.
in leishmaniasis: a focus on recent advances and challenges. Nanomaterials 2019. [184] Griesser J, Hetényi G, Kadas H, Demarne F, Jannin V, Bernkop-Schnürch A. Self-
12-1749. emulsifying peptide drug delivery systems: How to make them highly mucus
[149] Mirvakili SM, Langer R. Wireless on-demand drug delivery. Nat Electron permeating. Int J Pharm 2018;538:159–66.
2021;4:464–77. [185] Villar AMS, Naveros BC, Campmany ACC, Trenchs MA, Rocabert CB, Bellowa LH.
[150] Juliano RL. The delivery of therapeutic oligonucleotides. Nucleic Acids Res Design and optimization of self-nanoemulsifying drug delivery systems
2016;44(14):6518–48. https://doi.org/10.1093/nar/gkw236. (SNEDDS) for enhanced dissolution of gemfibrozil. Int J Pharm
[151] Tan X, Jia F, Wang P, Zhang K. Nucleic acid-based drug delivery strategies. J 2012;431:161–75.
Control Release 2020;10(323):240–52. https://doi.org/10.1016/j. [186] Porter CJH, Pouton CW, Cuine JF, Charman WN. Enhancing intestinal drug
jconrel.2020.03.040. solubilisation using lipid-based delivery systems. Adv Drug Deliv Rev
[152] Marques JT, Williams BR. Activation of the mammalian immune system by 2008;60:673–91.
siRNAs. Nat Biotechnol 2005;23:1399–405. [187] Kontogiannidou E, Meikopoulos T, Gika H, Panteris E, Vizirianakis IS, Müllertz A,
[153] He S, Ge Z, Zuo X, Fan C, Mao X. Dynamic regulation of DNA nanostructures by et al. In vitro evaluation of self-nano-emulsifying drug delivery systems
noncanonical nucleic acids. NPG Asia Mater 2021;13:1–2. (SNEDDS) containing room temperature ionic liquids (RTILs) for the oral
[154] Chandrasekaran AR. Nuclease resistance of DNA nanostructures. Nat Rev Chem delivery of amphotericin B. Pharmaceutics 2020;12:699.
2021;5(4):225–39. [188] Gursoy RN, Benita S. Self-Emulsifying Drug Delivery Systems (SEDDS) for
[155] Seeman NC. Nucleic acid junctions and lattices. J Theor Biol 1982;99:237–47. improved oral delivery of lipophilic drugs. Biomed Pharmacother
[156] Linko V, Kostiainen MA. Automated design of DNA origami. Nat Biotechnol 2004;58:173–82.
2016;34:826–7. [189] Larsen AT, Åkesson P, Juréus A, Saaby L, Abu-Reh R, Abrahamsson B, et al.
[157] Ge Z, Guo L, Wu G, Li J, Sun Y, Hou Y, et al. DNA origami-enabled engineering of Bioavailability of cinnarizine in dogs: effect of SNEDDS loading level and
ligand-drug conjugates for targeted drug delivery. Small 2020;16:1904857. correlation with cinnarizine solubilization during in vitro lipolysis. Pharm Res
[158] Wang W, Chen S, An B, Huang K, Bai T, Xu M, et al. Complex wireframe DNA 2013;30:3101–13.
nanostructures from simple building blocks. Nat Commun 2019;10:1–8. [190] Buya AB, Beloqui A, Memvanga PB, Préat V. Self-nano-emulsifying drug-delivery
[159] Jun H, Wang X, Bricker WP, Bathe M. Automated sequence design of 2D systems: From the development to the current applications and challenges in oral
wireframe DNA origami with honeycomb edges. Nat Commun 2019;10:1–9. drug delivery. Pharmaceutics 2020;12:1194.
[160] Popat A, Liu J, Lu GQ, Qiao SZ. A pH-responsive drug delivery system based on [191] Li, L.; Zhou, C.H.; Xu, Z.P. Self-Nanoemulsifying Drug-Delivery System. In
chitosan coated mesoporous silica nanoparticles. J Mater Chem Nanocarriers for Drug Delivery; Elsevier: Amsterdam, The Netherlands, 2019,
2012;22:11173–8. 421–449.
[161] Tian H, Chen J, Chen X. Nanoparticles for gene delivery. Small 2013;9:2034–44. [192] Cerpnjak K, Zvonar A, Gašperlin M, Vreˇcer F. Lipid-based systems as a promising
[162] Wu Y, Chen W, Meng F, Wang Z, Cheng R, Deng C, et al. Core-crosslinked pH- approach for enhancing the bioavailability of poorly water-soluble drugs. Acta
sensitive degradable micelles: a promising approach to resolve the extracellular Pharm 2013;63:427–45.
stability versus intracellular drug release dilemma. J Control Release [193] Memvanga PB, Coco R, Préat V. An oral malaria therapy: curcumin-loaded lipid-
2012;164:338–45. based drug delivery systems combined with β-arteether. J Control Release
[163] Park K. Controlled drug delivery systems: past forward and future back. J Control 2013;172:904–13.
Release 2014;190:3–8. [194] Syukri Y, Martien R, Lukitaningsih E, Nugroho AE. Novel self-nano emulsifying
[164] Chen W, Zhong P, Meng F, Cheng R, Deng C, Feijen J, et al. Redox and pH- drug delivery system (SNEDDS) of andrographolide isolated from Andrographis
responsive degradable micelles for dually activated intracellular anticancer drug paniculata Nees: characterization, in vitro and in vivo assessment. J Drug
release. J Control Release 2013;169:171–9. Delivery Sci Technol 2018;47:514–20.

19
A. Sultana et al. Medicine in Drug Discovery 15 (2022) 100134

[195] Makadia HA, Bhatt AY, Parmar RB, Paun JS, Tank HM. Self-nano emulsifying InIOP Conference Series: Materials Science and Engineering, 2011, 18, 192019 IOP
drug delivery system (SNEDDS): future aspects. Asian J Pharm Res 2013;3:21–7. Publishing.
[196] Dokania S, Joshi AK. Self-microemulsifying drug delivery system (SMEDDS)– [228] Paul W, Sharma CP. Bioceramics, towards nano-enabled drug delivery: a mini
challenges and road ahead. Drug Deliv 2015;22:675–90. review. Trends Biomater Artif Organs 2005;1:7–11.
[197] Patel D, Sawant KK. Self micro-emulsifying drug delivery system: formulation [229] Oliveira TM, Berti FCB, Gasoto SC, Schneider Jr B, Stimamiglio MA, Berti LF.
development and biopharmaceutical evaluation of lipophilic drugs. Curr Drug Calcium phosphate-based bioceramics in the treatment of osteosarcoma: drug
Deliv 2009;6:419–24. delivery composites and magnetic hyperthermia agents. Front Med Technol
[198] Visetvichaporn V, Kim KH, Jung K, Cho YS, Kim DD. Formulation of self- 2021;3:700266. https://doi.org/10.3389/fmedt.2021.700266.
microemulsifying drug delivery system (SMEDDS) by D-optimal mixture design [230] Sokolova V, Epple M. Biological and medical applications of calcium phosphate
to enhance the oral bioavailability of a new cathepsin K inhibitor (HL235). Int J nanoparticles. Chem Eur J 2021;27:7471–88.
Pharm 2020;573:118772. [231] Behrad Ghiasi, Yahya Sefidbakht and Maryam Rezaei, Hydroxyapatite for
[199] Adepu S, Ramakrishna S. Controlled drug delivery systems: current status and Biomedicine and Drug Delivery, In M. Rahmandoust and M. R. Ayatollahi
future directions. Molecules 2021;26:5905. https://doi.org/ (eds.), Nanomaterials for Advanced Biological Applications, Advanced Structured
10.3390/molecules2619590. Materials 104, Springer Nature Switzerland AG 2019 Pages 85-120, https://doi.
[200] Cao M, Wang Y, Hu X, Gong H, Li R, Cox H, et al. Reversible thermoresponsive org/10.1007/978-3-030-10834-2_4.
peptide–PNIPAM hydrogels for controlled drug delivery. Biomacromolecules [232] Shi H, Cheng Q, Yuan S, Ding X, Liu Y. Human serum albumin conjugated
2019;20:3601–10. nanoparticles for pH and redox-responsive delivery of a prodrug of cisplatin.
[201] Chang R, Tsai W. Photo-induced thermal-responsive nanogels for controlled drug Chemistry–A European Journal 2015;21:16547–54.
release. Front Bioeng Biotechnol Conference Abstract: 10th World Biomaterials [233] Kabanov AV, Vinogradov SV. Nanogels as pharmaceutical carriers: finite
Congress 2016. https://doi.org/10.3389/conf.FBIOE.2016.01.00807. networks of infinite capabilities. Angew Chem Int Ed 2009;48:5418–29.
[202] Vivek R, Babu VN, Thangam R, Subramanian KS, Kannan S. pH-responsive drug [234] Cuggino JC, Blanco ER, Gugliotta LM, Igarzabal CI, Calderón M. Crossing
delivery of chitosan nanoparticles as Tamoxifen carriers for effective anti-tumor biological barriers with nanogels to improve drug delivery performance. J
activity in breast cancer cells. Colloids Surf, B 2013;111:117–23. Control Release 2019;307:221–46.
[203] Lee S, Song SJ, Lee J, Ha TH, Choi JS. Cathepsin B-responsive liposomes for [235] Peng S, Wang H, Zhao W, Xin Y, Liu Y, Yu X, et al. Zwitterionic polysulfamide
controlled anticancer drug delivery in Hep G2 cells. Pharmaceutics 2020;12:876. drug nanogels with microwave augmented tumor accumulation and on-demand
[204] Kim H, Jang H, Kim B, Kim MK, Wie DS, Lee HS, et al. Flexible elastomer patch drug release for enhanced cancer therapy. Adv Funct Mater 2020;30:2001832.
with vertical silicon nanoneedles for intracellular and intratissue nanoinjection of [236] Li Y, Bui QN, Duy LT, Yang HY, Lee DS. One-step preparation of pH-responsive
biomolecules. Sci Adv 2018;4:eaau6972. polymeric nanogels as intelligent drug delivery systems for tumor therapy.
[205] Wang Z, Yang Y, Xu Z, Wang Y, Zhang W, Shi P. Interrogation of cellular innate Biomacromolecules 2018;19:2062–70.
immunity by diamond-nanoneedle-assisted intracellular molecular fishing. Nano [237] Kazemzadeh H, Mozafari M. Fullerene-based delivery systems. Drug Discovery
Lett 2015;15:7058–63. Today 2019;24:898–905.
[206] Wang Y, Yang Y, Yan L, Kwok SY, Li W, Wang Z, et al. Poking cells for efficient [238] Rezayat SM, Boushehri SV, Salmanian B, Omidvari AH, Tarighat S, Esmaeili S,
vector-free intracellular delivery. Nat Commun 2014;5:1–9. et al. The porphyrin–fullerene nanoparticles to promote the ATP overproduction
[207] Brotchie A. Drug delivery: ultrasound soothes the pain. Nat Rev Mater 2017;2:1. in myocardium: 25Mg2+-magnetic isotope effect. Eur J Med Chem
[208] Landhuis E. Ultrasound for the brain. Nature 2017;551(7679):257–9. 2009;44:1554–69.
[209] Couvreur P. Ultrasound-triggered pain relief. Nat Biomed Eng 2017;1:625–6. [239] Wan Y, Xu L, Zhuo N, Lu X. Retracted: A novel DNA sensor based on C60NPs-
[210] Shapiro MG, Goodwill PW, Neogy A, Yin M, Foster FS, Schaffer DV, et al. PAMAM-PtPNPs to detect VKORC1 gene for guiding rational clinical therapy
Biogenic gas nanostructures as ultrasonic molecular reporters. Nat Nanotechnol with Warfarin. Anal Chim Acta 2019;39:1009.
2014;9:311–6. [240] Venkatesan N, Yoshimitsu J, Ito Y, Shibata N, Takada K. Liquid filled
[211] Bourdeau RW, Lee-Gosselin A, Lakshmanan A, Farhadi A, Kumar SR, Nety SP, nanoparticles as a drug delivery tool for protein therapeutics. Biomaterials
et al. Acoustic reporter genes for noninvasive imaging of microorganisms in 2005;26:7154–63.
mammalian hosts. Nature 2018;553:86–90. [241] Pochkaeva EI, Podolsky NE, Zakusilo DN, Petrov AV, Charykov NA, Vlasov TD,
[212] O’Reilly MA, Chinnery T, Yee ML, Wu SK, Hynynen K, Kerbel RS, et al. et al. Fullerene derivatives with amino acids, peptides and proteins: from
Preliminary investigation of focused ultrasound-facilitated drug delivery for the synthesis to biomedical application. Prog Solid State Chem 2020;57:100255.
treatment of leptomeningeal metastases. Sci Rep 2018;8:1–8. [242] Liu D, Yang F, Xiong F, Gu N. The smart drug delivery system and its clinical
[213] Kong YL, Traverso G. Transmitting location. Nat Biomed Eng 2017;1:684–5. potential. Theranostics 2016;6:1306.
[214] Svirskis D, Travas-Sejdic J, Rodgers A, Garg S. Electrochemically controlled drug [243] Patravale V, Dandekar P, Jain R. Regulatory aspects of nanoparticulate drug
delivery based on intrinsically conducting polymers. J Control Release delivery systems. 2012:157–90.
2010;146:6–15. [244] Dave V, Sur S, Gupta N. Current framework, ethical consideration and future
[215] Maschietto M, Dal Maschio M, Girardi S, Vassanelli S. In situ electroporation of challenges of regulatory approach for nano-based products. Nanopharm Adv
mammalian cells through SiO2 thin film capacitive microelectrodes. Sci Rep Deliv Syst 2021:447–72.
2021;11(1-1). [245] Itani R, Tobaiqy M, Al FA. Optimizing use of theranostic nanoparticles as a life-
[216] Zaheer T, Pal K, Zaheer I. Topical review on nano-vaccinology: Biochemical saving strategy for treating COVID-19 patients. Theranostics 2020;10:5932.
promises and key challenges. Process Biochem 2021;100:237–44. [246] Ledford H. Hopes rise for coronavirus drug remdesivir. Nature 2020.
[217] Jeevanandam J., Danquah M.K. Nanosensors for better diagnosis of health. In: Pal [247] Cojocaru FD, Botezat D, Gardikiotis I, Uritu CM, Dodi G, Trandafir L, et al.
Kaushik, Gomes Fernando., editors. Micro and Nano Technologies, Nanomaterials designed for antiviral drug delivery transport across biological
Nanofabrication for Smart Nanosensor Applications. Elsevier; 2020, 187–228. barriers. Pharmaceutics 2020;12:171.
ISBN 9780128207024. [248] Zhu S, Huang AG, Luo F, Li J, Li J, Zhu L, et al. Application of virus targeting
[218] Kisby T, Yilmazer A, Kostarelos K. Reasons for success and lessons learnt from nanocarrier drug delivery system in virus-induced central nervous system disease
nanoscale vaccines against COVID-19. Nat Nanotechnol 2021;6:843–50. treatment. ACS Appl Mater Interfaces 2019;11:19006–16.
[219] Schüle S, Schulz-Fademrecht T, Garidel P, Bechtold-Peters K, Frieß W. [249] DiMasi JA, Feldman L, Seckler A, Wilson A. Trends in risks associated with new
Stabilization of IgG1 in spray-dried powders for inhalation. Eur J Pharm drug development: success rates for investigational drugs. Clin Pharmacol Ther
Biopharm 2008;69:793–807. 2010;87:272–7.
[220] Mahler HC, Müller R, Frieβ W, Delille A, Matheus S. Induction and analysis of [250] Alteri E, Guizzaro L. Be open about drug failures to speed up research. Nature
aggregates in a liquid IgG1-antibody formulation. Eur J Pharm Biopharm 2018;563:317–9. https://doi.org/10.1038/d41586-018-07352-7.
2005;59:407–17. [251] Zhou S. Bacteria synchronized for drug delivery. Nature 2016;536:33–4.
[221] Sou T, Meeusen EN, de Veer M, Morton DA, Kaminskas LM, McIntosh MP. New [252] Silva KC, Cezarino EC, Michelon M, Sato AC. Symbiotic microencapsulation to
developments in dry powder pulmonary vaccine delivery. Trends Biotechnol enhance Lactobacillus acidophilus survival. LWT 2018;89:503–9.
2011;29:191–8. [253] Ananthoji R, Eubank JF, Nouar F, Mouttaki H, Eddaoudi M, Harmon JP.
[222] Wang SH, Kirwan SM, Abraham SN, Staats HF, Hickey AJ. Stable dry powder Symbiosis of zeolite-like metal–organic frameworks (rho-ZMOF) and hydrogels:
formulation for nasal delivery of anthrax vaccine. J Pharm Sci 2012;101:31–47. Composites for controlled drug release. J Mater Chem 2011;21:9587–94.
[223] Velasquez LS, Shira S, Berta AN, Kilbourne J, Medi BM, Tizard I, et al. Intranasal [254] Brady E, Nielsen MW, Andersen JP, Oertelt-Prigione S. Lack of consideration of
delivery of Norwalk virus-like particles formulated in an in situ gelling, dry sex and gender in COVID-19 clinical studies. Nat Commun 2021;12:1–6.
powder vaccine. Vaccine 2011;29:5221–31. [255] Joung KI, Jung GW, Park HH, Lee H, Park SH, Shin JY. Gender differences in
[224] von Halling LC, Gibson B, van de Weert M, Boyd BJ, Rades T, Boisen A, et al. adverse event reports associated with antidiabetic drugs. Sci Rep 2020;10:1.
Spray dried cubosomes with ovalbumin and Quil-A as a nanoparticulate dry [256] Planelles B, Margarit C, Ballester P, Muriel J, Barrachina J, Ajo R, et al. Gender
powder vaccine formulation. Int J Pharm 2018;550:35–44. based differences, pharmacogenetics and adverse events in chronic pain
[225] Luczo JM, Bousse T, Johnson SK, Jones CA, Pearce N, Neiswanger CA, et al. management. Pharmacogenomics J 2020;20:320–8.
Intranasal powder live attenuated influenza vaccine is thermostable, [257] Cirillo D, Catuara-Solarz S, Morey C, Guney E, Subirats L, Mellino S, et al. Sex and
immunogenic, and protective against homologous challenge in ferrets. npj gender differences and biases in artificial intelligence for biomedicine and
Vaccines 2021;6:1–8. healthcare. npj Digital Med 2020;3. 1–1.
[226] Sokolova V, Epple M. Bioceramic nanoparticles for tissue engineering and drug [258] Eichler HG, Enzmann H, Rasi G. Added therapeutic benefit and drug licensing.
delivery. InTissue Engineering Using Ceramics and Polymers, 2014, 633-647. Nat Rev Drug Discovery 2019;18:651–2.
Woodhead Publishing. [259] Zhang Y, Ge J. Green nanoparticles for oligonucleotide delivery. Gene Ther
[227] Loca D, Locs J, Salma K, Gulbis J, Salma I, Berzina-Cimdina L. Porous 2020;27:535–6.
hydroxyapatite bioceramic scaffolds for drug delivery and bone regeneration.

20
A. Sultana et al. Medicine in Drug Discovery 15 (2022) 100134

[260] Mohammadinejad R, Madamsetty VS, Kumar A, Varzandeh M, Dehshahri A, [264] Chen T, Wu W, Xiao H, Chen Y, Chen M, Li J. Intelligent drug delivery system
Zarrabi A, et al. Electrospun nanocarriers for delivering natural products for based on mesoporous silica nanoparticles coated with an ultra-pH-sensitive
cancer therapy. Trends Food Sci Technol 2021;118:887–904. gatekeeper and poly (ethylene glycol). ACS Macro Lett 2016;5:55–8.
[261] Arun A, Malrautu P, Laha A, Luo H, Ramakrishna S. Collagen nanoparticles in [265] Wang X, Shao J, Abd El Raouf M, Xie H, Huang H, Wang H, et al. Near-infrared
drug delivery systems and tissue engineering. Appl Sci 2021;11:11369. light-triggered drug delivery system based on black phosphorus for in vivo bone
[262] Alvarez-Lorenzo C, Concheiro A. Intelligent drug delivery systems: polymeric regeneration. Biomaterials 2018;179:164–74.
micelles and hydrogels. Mini Rev Med Chem 2008;8:1065–74. [266] Sharma R, Singh D, Gaur P, Joshi D. Intelligent automated drug administration
[263] Jain A, Mohanty PK. Self regulatory drug transport: an intelligent drug delivery and therapy: future of healthcare. Drug Deliv Transl Res 2021;11:1–25.
system. Ars Pharm 2018;59:173–83.

21

You might also like