Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 4152 word(s) 4152 2021-11-03 10:34:52 |
2 format correction Meta information modification 4152 2021-11-11 03:52:16 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Ong, Y.S. Lipid-Based Nanoparticles for Encapsulation of Anticancer Alkaloids. Encyclopedia. Available online: https://encyclopedia.pub/entry/15888 (accessed on 08 July 2024).
Ong YS. Lipid-Based Nanoparticles for Encapsulation of Anticancer Alkaloids. Encyclopedia. Available at: https://encyclopedia.pub/entry/15888. Accessed July 08, 2024.
Ong, Yong Sze. "Lipid-Based Nanoparticles for Encapsulation of Anticancer Alkaloids" Encyclopedia, https://encyclopedia.pub/entry/15888 (accessed July 08, 2024).
Ong, Y.S. (2021, November 11). Lipid-Based Nanoparticles for Encapsulation of Anticancer Alkaloids. In Encyclopedia. https://encyclopedia.pub/entry/15888
Ong, Yong Sze. "Lipid-Based Nanoparticles for Encapsulation of Anticancer Alkaloids." Encyclopedia. Web. 11 November, 2021.
Lipid-Based Nanoparticles for Encapsulation of Anticancer Alkaloids
Edit

Alkaloids are natural products that possess numerous pharmacological activities and have been exploited effectively to treat cancer. However, the clinically approved anticancer alkaloids are generally limited by serious side effects due to their lack of specificity to cancer cells, indiscriminate tissue distribution and toxic formulation excipients. Lipid-based nanoparticles represent the most effective drug delivery system concerning clinical translation owing to their unique appealing characteristics for drug delivery.

alkaloid anticancer lipid-based nanoparticles liposome solid lipid nanoparticle nanostructured lipid carriers

1. Introduction

Cancer ranks as the leading cause of morbidity and mortality in the world with an estimated 19.3 million new cases and 9.9 million deaths reported in 2020 [1]. Although stupendous advances have been made in understanding the molecular underpinnings and genomic landscape of cancers, the oncologic outcomes remain poor. Current treatments of cancer include surgery, radiation therapy and chemotherapy [2][3][4]. However, the administration of anti-cancer drugs, including chemotherapeutic drugs, biologic agents and immunotherapeutic drugs using the conventional methods, has been hindered by various pharmacological issues, including toxicities, unsatisfactory therapeutic efficacy and drug resistance [5][6]. These unsatisfactory oncologic outcomes have revitalized the interest in natural product-derived anticancer agents.
Bioactive natural products have been serving as the primary source of medicines by numerous cultures around the world over the past millennia [7]. With the rise of modern scientific approaches, the past century has witnessed a surge of highly active compounds derived from natural products and their derivatives with a precise mode of actions for the treatment of a myriad of diseases. Natural products have gained great interest due to their vast scaffold diversity and structural complexity unrivaled by current synthetic drugs [8]. An analysis of all FDA-approved small-molecule drugs from 1981 to 2014 revealed that approximately 51% were natural products and their derivatives, and about 80% of anti-cancer small-molecule drugs were natural products and their derivatives [9]. Several classes of natural products have been identified, including terpenoid, polyketide, phenylpropanoid and alkaloid [10].
Alkaloid is a class of naturally occurring nitrogen containing heterocyclic organic compounds with a wide range of pharmacological activities, often considered privileged structures in drug discovery [10][11]. Since the commercialization of the first alkaloid morphine in 1826, numerous alkaloids have been isolated and exploited effectively for the betterment of mankind. Today, alkaloid drugs have been approved by the FDA for the treatment of cancer, Alzheimer’s disease, Parkinson’s disease, migraine, pain control, erectile dysfunction, heart failure and many more [12][13]. Alkaloids have demonstrated wide-spectrum anticancer activity by inhibiting topoisomerase I and suppressing microtubule dynamics [14][15]. The most notable anticancer alkaloid drugs that continue to maintain the palpable significance in clinical practice include paclitaxel, docetaxel, vincristine, vinblastine, irinotecan and topotecan. However, the administration of anticancer alkaloids has generally been limited by serious side effects due to their lack of specificity to cancer cells, indiscriminate tissue distribution and toxic formulation excipients [16][17][18]. These limitations prompted unceasing investigational efforts to develop effective and safe nanoformulations and improve oncologic outcomes.
Cancer is a disease where the adequacy of delivery of extremely potent yet toxic chemotherapeutic drugs can result in either efficacious responses or serious morbidity [19]. To mitigate these limitations, tailor-designed nanomedicines have emerged as a promising strategy for cancer treatment owing to their improved pharmacokinetic properties, therapeutic efficacy, specific targeting of tissues and minimized adverse effects [20]. Furthermore, the use of nanotechnology allows drugs to traverse biological barriers such as the blood-brain barrier [19][21]. Among all the different classes of nanocarriers, lipid-based nanoparticles represent the most established and effective drug delivery system concerning clinical translation, with multiple formulations having already obtained U.S. Food and Drug Administration (FDA) approval for clinical use [20]. Lipid-based nanoparticles have received great attention due to their unique, appealing characteristics for drug delivery, including (1) excellent biocompatibility and biodegradability; (2) improved solubility and stability of difficult-to-deliver drugs, including both hydrophobic and hydrophilic drugs; (3) enhanced therapeutic index by improving efficacy and reducing toxicity; (4) versatility which allows chemical modifications and surface coatings and (5) ability of co-deliver two different anticancer drugs to enable precise spatiotemporal multi-drug treatment [22][23][24][25]. These advantages of lipid-based nanoparticles have been exploited effectively in enhancing the efficacy and reducing the toxicity of anticancer alkaloids, the most exceptional of which are Onivyde (liposomal irinotecan), Marqibo (liposomal vincristine) and Lipusu (liposomal paclitaxel), which have received regulatory approval for clinical use [26]. These uplifting translational successes have motivated an exponential increase in research investigating the potential and effectiveness of encapsulating anticancer alkaloids in different lipid-based nanoparticles. Nevertheless, despite the stupendous therapeutic potential demonstrated by nanomedicines in pre-clinical studies, there are still many shortcomings to be solved.

2. Nanotechnology

The prefix “nano” comes from ancient Greek which represents “dwarf” [27]. One nanometer (nm) is an international System of Units that equals to one billionth of a meter. According to the National Nanotechnology Initiative (NNI), nanotechnology refers to the comprehension and manipulation of matter at atomic or molecular levels between 1 to 100 nm, where unique phenomena facilitate the novel applications [28]. It involves various scientific disciplines (i.e., engineering, technology, medicine, chemistry, physics, biology or a combination of these disciplines) [29]. Nanotechnology has been utilized in numerous medical-related fields including magnetic resonance imaging (MRI), hyperthermic destruction of tumor, proteins detection, diagnosis, pharmacological research, cells and biological molecules purification. Nowadays, numerous nanoparticles have been studied and developed for clinical use including liposomes, nanocapsules, nanorods, nanowires, nanospheres, nanoshells, nanotubes, nanopores and dendrimers [27][30][31].
In general, nanomaterials can be divided into four different categories which are carbon-based, inorganic-based, organic-based and composite-based nanomaterials. Carbon-based nanomaterials can be found in morphologies such as sphere-shaped, ellipsoid-shaped, tube-shaped or horn-shaped [32]. These nanomaterials can further be classified into graphene quantum dots (0-D), carbon nanotubes (1-D) and graphene (2-D) based on their dimensions, where 0-D refers to no dimension, 1-D refers to one dimension and 2-D refers to two dimensions at nanoscale [33][34]. Inorganic-based nanomaterials comprise metal-based and metal oxide-based nanoparticles. Metal-based nanoparticles are comprised of pure metal nanoparticles (i.e., iron, magnesium, zinc, platinum, titanium, copper, gold, silver and alginate nanoparticles). Metal-based nanoparticles can be bound to oxygen, becoming metal oxide nanomaterials (i.e., zinc oxide, silver oxide, etc.) [35][36]. Organic-based nanomaterials are mainly made of organic matter except inorganic and carbon-based nanomaterials. These organic-based nanomaterials can be transformed into liposomes, micelles, dendrimers and polymer nanoparticles that are very useful in drug delivery through noncovalent interactions [36][37]. Composite-based nanomaterials consist of several phases, with one of the phases on a nanoscale dimension that merges different nanoparticles together with enormous or complicated materials, for example hybrid nanofibers and metal-organic frameworks. A composite of these nanomaterials can be a mixture of any polymer, ceramic or metal materials with any organic-based, inorganic-based, metal-based or carbon based nanomaterials [36]. These nanomaterials have potency to revolutionizes the manner at which diseases such as cancer are diagnosed and treated.

3. Alkaloid

Alkaloids are ubiquitous in nature. They are mostly found in plants, and can also be produced by terrestrial animals, marine organisms, microorganisms such as bacteria, fungi and insects. Approximately 20% of plant species contain alkaloids, most of which are biosynthetically derived from amino acids lysine (Lys), ornithine (Orn), tryptophan (Trp), tyrosine (Tyr) and phenylalanine (Phe) [38].
Alkaloid is a class of naturally occurring heterocyclic organic compounds that contain a nitrogen atom. With over 20,000 structurally characterized members, alkaloids remain one of the most medicinally important classes of compounds with a wide range of pharmacological activities, often considered privileged structures in drug discovery [10][11]. In fact, the first naturally derived pure medicine was morphine, an alkaloid isolated from opium poppy in year 1805 and it was commercialized by Merck in 1826 [13]. Since then, numerous alkaloids have been isolated and exploited effectively for the betterment of mankind.
Due to its vast structural diversity and widespread distribution in nature, several classification systems have been used to classify alkaloids, including chemical classification, taxonomic classification, pharmacological classification and biosynthetic classification, each with their own strengths and limitations. In this review, we have adopted the chemical classification to classify the alkaloids based on their chemical structures as this is the most established classification scheme for alkaloids. On this classification basis, the main classes of alkaloids in our review are indole, quinoline, isoquinoline, pyrrolidine, pyridine, piperidine, tropane, indolizidine, terpenoid, purine, imidazole and steroidal alkaloids. Table 1 summarizes different types of medicinally significant alkaloids according to the chemical classification.
Table 1. Medicinally Significant Alkaloids According to the Chemical Classification.

Class

Drugs

Molecular Formula

Origin

Indication/Uses

Cancers 13 05346 i001

Indole

Vincristine

Cancers 13 05346 i002

C46H56N4O10

Catharanthus roseus

Anticancer

Vinblastine

Cancers 13 05346 i003

C46H58N4O9

Vinorelbine

Cancers 13 05346 i004

C45H54N4O8

Vincamine

Cancers 13 05346 i005

C21H26N2O3

Vinca minor

Primary degenerative and vascular dementia

Physostigmine

Cancers 13 05346 i006

C15H21N3O2

Physostigma venenosum

Glaucoma

Ajmaline

Cancers 13 05346 i007

C20H26N2O2

Rauvolfia serpentina

Anti-arrhythmic

Ajmalicine

Cancers 13 05346 i008

C21H24N2O3

Anti-hypertensive

Reserpine

Cancers 13 05346 i009

C33H40N2O9

Rauvolfia serpentina

Anti-hypertensive

Yohimbine

Cancers 13 05346 i010

C21H26N2O3

Erectile dysfunction

Strychnine

Cancers 13 05346 i011

C21H22N2O2

Strychnos nux-vomica

Convulsant

Mitragynine

Cancers 13 05346 i012

C23H30N2O4

Mitragyna speciosa

Stimulant, analgesic

Psilocin

Cancers 13 05346 i013

C12H16N2O

Psilocybe cubensis

Hallucinogen

Psilocybin

Cancers 13 05346 i014

C12H17N2O4P

Ephedrine

Cancers 13 05346 i015

C6H5CH(OH)CH (CH3)NHCH3

Ephedra sinica

Bronchial asthma

Cancers 13 05346 i016

Quinoline

Irinotecan

Cancers 13 05346 i017

C33H38N4O6

Catharanthus roseus

Anticancer

Topotecan

Cancers 13 05346 i018

C23H23N3O5

Colchicine

Cancers 13 05346 i019

C22H25NO6

Colchicum autumnale

Gout

Quinidine

Cancers 13 05346 i020

C20H24N2O2

Cinchona officinalis

Anti-arrhythmic

Quinine

Cancers 13 05346 i021

C20H24N2O2

Anti-malarial

Cinchonine

Cancers 13 05346 i022

C19H22N2O

Cinchonidine

Cancers 13 05346 i023

C19H22N2O

Cancers 13 05346 i024

Isoquinolines

Morphine

Cancers 13 05346 i025

C34H40N2O10S

Papaver somniferum

Analgesic

Codeine

Cancers 13 05346 i026

C18H21NO3

Heroine

Cancers 13 05346 i027

C21H23NO5

Apomorphine

Cancers 13 05346 i028

C17H17NO2

Parkinson’s disease

Noscapine

Cancers 13 05346 i029

C22H23NO7

Anti-tussive

Trabectedin

Cancers 13 05346 i030

C39H43N3O11S

Ecteinascidia turbinata

Anticancer

Berberine

Cancers 13 05346 i031

C20H18ClNO4

Coptis chinensis

Antimicrobial

Tubocurarine

Cancers 13 05346 i032

C37H41ClN2O6

Chondrodendron tomentosum

Skeletal muscle relaxant

Atracurium

Cancers 13 05346 i033

C53H72N2O12

Leontice leontopetalum

Tetrandrine

Cancers 13 05346 i034

C38H42N2O6

Stephania tetrandra

Anti-arrhythmic

Galantamine

Cancers 13 05346 i035

C17H21NO3

Galanthus nivalis

Alzheimer’s disease

Sanguinarine

Cancers 13 05346 i036

C20H14NO4

Sanguinaria canadenis

Antibacterial, antiplaque

Papaverine

Cancers 13 05346 i037

C20H21NO4

Papaver somniferum

Vasodilator

Cancers 13 05346 i038

Pyrrolidines

Hygrine

Cancers 13 05346 i039

C8H15NO

Erythroxylon coca

Laxative, diuretic

Cuscohygrine

Cancers 13 05346 i040

C13H24N2O

Stachydrine

Cancers 13 05346 i041

C7H14NO2

Stachys tuberifera

Neuroprotectant

Cancers 13 05346 i042

Pyridines

Arecoline

Cancers 13 05346 i043

C8H13NO2

Areca catechu

Muscarinic agonist

Ricinine

Cancers 13 05346 i044

C8H8N2O2

Ricinus communis

Insecticide

Trigonelline

Cancers 13 05346 i045

C7H7NO2

Trigonella foenum-graecum

Antidiabetic

Nicotine

Cancers 13 05346 i046

C10H14N2

Nicotiana tabacum

Smoking cessation

Cancers 13 05346 i047

Piperidine

Piperine

Cancers 13 05346 i048

C17H19NO3

Piper nigrum

Anticancer

Piperlongumine

Cancers 13 05346 i049

C17H19NO5

Piper longum

Pipernonaline

Cancers 13 05346 i050

C21H27NO3

Antifungal

Cancers 13 05346 i051

Tropanes

Atropine

Cancers 13 05346 i052

C17H23NO3

Atropa belladonna

Anticholinergic

Cocaine

Cancers 13 05346 i053

C17H21NO4

Erythroxylum coca

Local anaesthetic

Hyoscyamine

Cancers 13 05346 i054

C17H23NO3

Atropa belladonna, Hyoscyamus niger

Anticholinergic

Hyoscine

Cancers 13 05346 i055

C17H21NO4

Atropa belladonna

Motion sickness

Cancers 13 05346 i056

Indolizidine

Swainsonine

Cancers 13 05346 i057

C8H15NO3

Swainsona canescens

Anticancer

Castanospermine

Cancers 13 05346 i058

C8H15NO4

Castanospermum australe

Antiviral

Securinine

Cancers 13 05346 i059

C13H15NO2

Securinega suffruticosa

Neuroprotection

Tylophorine

Cancers 13 05346 i060

C24H27NO4

Tylophora indica

Anticancer

Lycorine

Cancers 13 05346 i061

C16H17NO4

Clivia miniata

Cancers 13 05346 i062

Terpenoids

Paclitaxel

Cancers 13 05346 i063

C47H51NO14

Taxus brevifolia

Docetaxel

Cancers 13 05346 i064

C43H53NO14

Taxus baccata

Cancers 13 05346 i065

Purine

Caffeine

Cancers 13 05346 i066

C8H10N4O2

Coffee arabica

CNS stimulant

Theobromine

Cancers 13 05346 i067

C7H8N4O2

Theobroma cacao

Cardioprotectant

Theophylline

Cancers 13 05346 i068

C7H8N4O2

COPD and asthma

Cancers 13 05346 i069

Imidazole

Pilocarpine

Cancers 13 05346 i070

C11H16N2O2

Pilocarpus microphyllus

Glaucoma

Epiisopiloturine

Cancers 13 05346 i071

C₁₆H₁₈N₂O₃

Anthelmintic

Cancers 13 05346 i072

Steroidal

Pancuronium

Cancers 13 05346 i073

C35H60N2O4

Malouetia bequaertiana

Skeletal muscle relaxant

Conessine

Cancers 13 05346 i074

C24H40N2

Holarrhena antidysenterica

Antidysenteric

Solanidine

Cancers 13 05346 i075

C27H43NO

Solanum tuberosum

Anticancer

Tomatidine

Cancers 13 05346 i076

C27H45NO2

Lycopersicon esculentum

Today, alkaloid drugs have been approved by the FDA for the treatment of cancer, Alzheimer’s disease, Parkinson’s disease, migraine, pain control, erectile dysfunction, heart failure and many more [12]. Alkaloids have been widely utilized in various solid tumors and hematological malignancies as a monotherapy or in combination with other chemotherapeutic drugs [39][40][41][42][43]. Thus, much effort has been devoted to elucidate and decipher the mechanism of action of these anticancer alkaloids.

4. Lipid-Based Nanoparticles for Encapsulation of Anticancer Alkaloids

Considering all the foregoing hints, nanocarriers could be a potential strategy to overcome the limitations of alkaloids. Nanocarriers are around 5 to 200 nm in size and can be used in a wide range of applications [44]. They can be categorized into various types (e.g., organic, inorganic, polymeric, biological, lipid-based nanocarriers) according to their physical properties, chemical properties, morphology and size. Among the carriers, lipid-based nanocarriers offer an alternative to solubilize, encapsulate and deliver alkaloids in a programmed manner to enhance their water solubility, bioavailability and anticancer efficacy [25]. Examples of lipid-based nanocarriers are liposomes, solid lipid nanoparticles (SLN) and nanostructured lipid-carriers (NLC) (Figure 1). These drug carriers are made up of biocompatible lipids triglycerides, cholesterol and phospholipids in which most of them are derivatized based on or extracted from natural sources, resulting in their excellent biodegradability and biocompatibility [45]. Excipients use in lipid carriers such as cholesterol, PEG and phosphatidylcholine have established toxicology data and safety profiles for their use in pharmaceutical products, further strengthening their potential as the ideal drug delivery system [46]. Lipid-based nanoparticles with an average size of 100 nm and have longer circulation half-lives which enhance their propensity to extravasate through vascular fenestrations of tumors’ vasculature, thus enhancing the potency of anticancer agents [47]. However, it is relatively difficult to prepare such small-sized lipid-based nanoparticles. In this regard, a “top down” size reduction approach that requires high energy input (e.g., sonication) and a “bottom up” method that produces nanoparticles by lipid condensation from solution can be used to solve this problem [48]. As far as we know, clinically approved cancer nanomedicines that utilize lipid-based nanocarriers as drug delivery agents have particle sizes larger than 80 nm, for example, Doxil80–100 nm), Marqibo (100 nm) and Abraxane (130 nm) [49][50][51]. Numerous studies have been conducted to encapsulate alkaloids into lipid nanocarriers.
Figure 1. Classification of Lipid-based Nanocarriers.

4.1. Liposome

Liposomes are one of the extensively studied lipid vesicles, which are made up of phospholipids and an aqueous medium. Lipid vesicles are formed when lipids interact with the aqueous medium, where the lipid hydrophilic head group envelops the aqueous core along with exposure of the hydrophilic tail group to the external medium. Owing to this distinct structural property, drugs can be entrapped in the lipid bilayer or loaded in the internal aqueous core of liposomes depending on their hydrophilicity, for example, Doxil and Onivyde [52]. The first nano-based formulations approved by the FDA for cancer treatment are liposomal anticancer drugs. Doxil was the first doxorubicin-loaded liposomal drug which received FDA approval in 1995 to be used in the treatment of AIDS-related multiple myeloma and Kaposi sarcoma due to its lower cardiotoxicity and higher efficacy as compared to free doxorubicin alone. The clinical success of Doxil further established the potential of liposomes in drug delivery, and many promising liposomal formulations are currently being scrutinized intensively in clinical studies [52][53]. Reduced dose-limiting toxicities, improved undesirable pharmacokinetics and drug solubility are the primary objectives in liposomal development to deliver alkaloids. Liposomes’ stability is one of the major issues in the development of liposomal alkaloids [45]. The drug to lipid ratio and lipid composition have to be taken into consideration in preparing physically stable liposomal alkaloids. Neutral zwitterionic lipids are the most commonly used lipids in preparing liposomes such as phosphatidylcholine. Phospholipids and cholesterol can be added to enhance stability, reduce aggregation and improve permeability of drugs [54][55]. Introduction of the PEGylation approach in the early nineties was shown to prolong the circulation time of liposome significantly due to steric stabilization of vesicles [56]. Likewise, incorporation of PEG-modified lipids was found to enhance delivery and cytotoxicity of paclitaxel liposome to human cancer cells [57]. However, PEG, which is hydrophobic in nature, was later found to lower the polarity of the aqueous matrix and destabilize the liposomes, leading to rapid drug leakage. To overcome the issue, poly(zwitterionic) polymers such as poly[2-(methacryloyloxy)ethyl phosphorylcholine) (PMPC) [58] and poly(carboxybetaine) (PCB) [59] were introduced to replace PEG. That said, liposomes have disadvantages including limited drug loading capacity due to space available in liposomal lipid membranes, inadequate control of drug release, reproducibility issues and stability issues [60]. The drug-loading capacity of liposome can be solved by a remote loading approach where the drug will be added to preformed liposomes via pH gradient or ion gradient competent to create a pH gradient [61].
In 2014, liposomes were used to co-encapsulate doxorubicin and a contrast agent (Magnevist) or hyperthermic agent (Fe3O4) [62][63]. Doxorubicin has also been encapsulated into ultrasound-sensitive liposomes in which the drug release can be facilitated by sonication that degrades liposomes, co-encapsulated with curcumin in liposomes to enhance anti-tumor efficacy of doxorubicin in the C26 colon cancer cell line, encapsulated in arginine-penetrating peptides/PEG modified liposomes to decrease in vitro cytotoxicity and enhance delivery of doxorubicin for the treatment of ovarian cancer [64][65][66]. In addition, incorporation of docetaxel in anacardic acid and PEG-modified liposomes have proven to stabilize docetaxel [67]. Many different liposomes and modified liposomes were designed to carry a variety of anticancer agents, such as paclitaxel and 5-fluorouracil [68].
Liposomal preparation requires a cryogenic atmosphere brought on to the introduction of vesicular drug delivery systems using non-ionic surfactants. It was named niosome consisting of either uni- or multi- lamellar vesicles. Niosome was first introduced in the cosmetic industry and its potential application in drug delivery was only discovered thenceforth [69]. The unique structure of niosome allows it to encapsulate both hydrophilic materials in vesicular aqueous core and lipophilic materials in the bilayer domain. It is composed of non-ionic surfactants with cholesterol and can increase the size and provide charge to vesicles, therefore, enhancing the entrapment efficiency of noisome. Niosome has a similar structure as liposomes and it is expected to be a better delivery system than liposomes due to its stability, cost and entrapment efficiency [70][71]. Nevertheless, only few niosome formulations are tested in clinical trials, and no formulations are commercially marketed heretofore owing to their low efficacy [72]. Ethosome is also a modified version of classical liposomes and is mainly composed of phospholipids, ethanol and water in which the concentration of ethanol is relatively high, differentiating it from other vesicular carriers. High ethanol constituents of ethosome improve skin permeability by releasing the encapsulated materials into deeper layers and systemic circulation [73][74]. For transdermal delivery of drugs, ethosome is superior over classical liposomes due to its higher entrapment efficiency, more negative zeta potential and smaller size [75][76]. Despite having these superiorities, only few ethosome formulations enter clinical trials due to limitations such as low yield and suitability to carry potent drugs only but not drugs which require a high concentration of blood [73].

4.2. Micelles

Micelles are colloidal systems formed through self-assembly of amphiphilic molecules. They can be further classified into polymeric micelles, lipid micelles and lipid polymeric hybrid micelles based on types of amphiphilic molecules. Amphiphilic molecules in lipid micelles are normally small-molecule surfactants. Unlike liposomes having a lipid bilayer, lipid micelles have a monolayer with an outer hydrophilic corona enclosing an inner hydrophobic core form by hydrophobic acyl chains [77]. Critical micelle concentration (CMC) refers to concentration of surfactants above which micelles form. It is an important surfactants parameter to consider in designing micelles [45]. Enhancing solubility of drugs is the primary objective of designing micelles as drug nano-carriers. Hydrophobic drugs such as docetaxel and paclitaxel are carried in the lipophilic core of micelles [77]. Nevertheless, lipid micelles possess major limitations: unstable in bloodstream as dissociation occurred upon dilution below the micelle forming concentration (CMC) and limited interior hydrophobic space influencing the loading capacity of drugs. In order to stabilize the micelles by reinforcing weak intermolecular interactions, several strategies such as the formation of covalent crosslinking (for i.e., shell crosslinking) and non-covalent crosslinking (for i.e., diblock copolymers) can be used [78]. Lipid micelle has been used to deliver various drugs including paclitaxel, doxorubicin and camptothecin in the preclinical stage [79][80][81]. Cabral and co-workers identified that small polymeric micelles are suitable in delivering drug to the tumor site due to their favorable size range between 30 to 100 nm which penetrates well in highly permeable tumors [48]. However, they are not up to the market until today due to insufficient cellular interaction with tumor cells for cellular uptake and poor physical stability in vivo [82]. Even though both micelles and vesicles were formulated in the same principle whereby the lipid molecules reorganized and clustered together in an aqueous solution, the lipid layers were formed differently depending on their shapes. Lipid micelles were formed by wedge-shaped lipid molecules with the hydrophobic tails facing inwards, whereas the vesicles were formed by the cylinder-shaped phospholipid molecules with the hydrophobic tails sandwiched between the hydrophilic head groups [83].

4.3. Solid Lipid Nanoparticles

Comparing the lipid-based nanocarriers discussed, solid lipid nanoparticles (SLN) represent a colloidal drug delivery system with an external aqueous phase and internal lipid phase introduced in the early 1990s. They are composed of a combination of various solid lipids such as waxes and fatty acids, as well as mono-, di- and triacylglycerols that form a lipid matrix entrapping drugs or other hydrophobic materials. Beneficial properties of SLN, such as cost effectiveness, non-toxicity, ease of preparation, controlled drug release, good system stability and provision of target-specific effects, made them outstrip other carriers when they were first introduced [84][85]. On top of that, use of IV acceptable solid lipids (e.g., phospholipids, glycerides) and surfactants (e.g., poloxamer 188, lecithin, tween-80) makes SLN a versatile platform for drug delivery readily to translate into clinical application [84]. SLN-based formulations have shown a substantial enhancement in the anti-tumor efficacy of hydrophobic drugs. For example, resveratrol-SLN have greater inhibitory effects against the proliferation, invasion and migration of breast cancer cells than the free drug alone; talazoparib-SLN improve the therapeutic index against triple-negative breast cancer by overcoming homologous recombination-mediated resistance and talazoparib toxicity [86]. However, the highly organized crystalline structure of solid lipids leaves a limited room for drug incorporation which contributes to low drug capacity and drug expulsion during storage [87]. However, the highly organized crystalline structure of solid lipids leaves a limited room for drug incorporation which contributes to a low drug-loading capacity and drug expulsion during storage [87]. To overcome this problem, Muller and colleagues have come out with a novel lipid delivery system called nanostructured lipid carriers (NLC) [88].

4.4. Nanostructured Lipid Carriers

NLC are a modified version of conventional SLN by incorporating liquid lipids with a solid lipid. The room between crystal imperfections and fatty acid chains of NLC allows more drug accommodation. Moreover, certain drugs are more soluble in liquid lipids than solid lipids [89]. Hence, NLC as the second generation lipid carriers after SLN reduce drug expulsion during storage and enhance the drug-loading capacity [87]. NLC can be prepared using various surfactants and co-surfactants, allowing them to be formulated for various administration routes (e.g., parenteral, oral, topical, ocular, nasal) to deliver drugs and active substances for biochemical, cosmetic and pharmaceutical purposes [89]. NLC possess favorable properties of low toxicity and controlled drug release, and provide target-specific effects and a high drug load for both hydrophilic and hydrophobic agents, making them outstrip SLN when they were introduced [84][90]. NLC-based formulations have been studied extensively for the delivery of anti-tumor agents. For example, 6-gingerol-NLC has a greater water solubility and oral bioavailability than the free drug alone, penetrating peptides, and hyaluronic-acid-modified artesunate-NLC has a better cell-membrane-penetrating ability against hepatic cancer; thymoquinone-NLC and fluvastatin-NLC have an improved anti-tumor efficacy against hepatic cancer and prostate cancer, respectively [91].
Lipid-based delivery systems attracted a great deal of attention in the past decades as a strategy to enhance anticancer efficacy and overcome delivery barriers and the therapeutic index of various drugs, especially alkaloids. Myriads of preclinical studies have been focused on the use of lipid carriers to deliver alkaloids due to their potential in cancer treatment. Table 2 summarizes alkaloids that have been encapsulated in lipid-based nanocarriers for cancer treatment. Some of them utilized vesicular systems while others used lipid-particulate systems.
Table 2. Alkaloids that have been Successfully Encapsulated in Lipid-based Nanoparticles.

Class

Compound

Type of Lipid Carrier

Type of Cancer

In Vitro/In Vivo

Cell Type/Animal Model

Ref.

Terpenoids

Docetaxel

Cancers 13 05346 i077

Liposome

Breast

in vitro; in vivo

MCF-7/4T1 xenograft mice

[92]

in vivo

MDA-MB-435/LCC6 xenograft mice

[93]

in vitro

MCF-7

[94]

Lung

in vivo

A549 xenograft rat

[95]

in vitro

A549

[96]

in vitro

A549

[94]

Liver

in vitro

HepG2

[94]

Melanoma

in vitro; in vivo

B16F10/B16-F10 xenograft mice

[97]

Micelle

Breast

in vitro

MCF-7

[98]

in vitro

MCF-7

[99]

Lung

in vitro

A549

[99]

Niosome

Breast

in vitro; in vivo

MDA-MB-s31 and MCF-7

[100]

NLC

Liver, Ovarian, Lung, Melanoma

in vitro; in vivo

HepG2, SKOV3, A549, B16 cells

[101]

Paclitaxel

Cancers 13 05346 i078

Liposome

Breast

in vitro; in vivo

4T1 xenograft mice

[102]

in vivo

ICR male mice

[103]

Lung

in vivo

A549 xenograft mice

[104]

in vitro; in vivo

A549

[105]

NLC

Breast, Ovarian

in vitro

MCF-7, SKOV3

[106]

Ethosome

Squamous Cell Carcinoma

in vitro

DJM-1

[107]

Micelle

Glioma

in vitro; in vivo

C6/C6 xenograft rat

[108]

Indole

Vincristine

Cancers 13 05346 i079

Liposome

Acute Lymphoblastic Leukemia

in vivo

Namwala xenograft mice

[49]

Brain Glioma

in vitro, in vivo

Glioma bearing mice

[109]

Nasopharyngeal cancer

in vitro, in vivo

KB/KBv200 xenograft mice

[110]

SLN

Breast

in vitro; in vivo

MDA-MB-231

[111]

NLC

Breast

in vitro

MCF-7

[112]

Vinblastine

Cancers 13 05346 i080

Liposome

Non-Small Cell Lung

in vitro; in vivo

LLT/LLT xenograft mice

[113]

Niosome

Lung

in vitro; in vivo

TC-1/TC-1 xenograft mice

[114]

Vinorelbine

Cancers 13 05346 i081

Micelle

Breast

in vitro

MCF-7

[115]

Quinoline

Topotecan

Cancers 13 05346 i082

Liposome

Lung and Adenocarcinoma

in vitro

LLC

[116]

Breast

in vitro; in vivo

MCF-7/MCF-7 xenograft mice

[117]

SLN/NLC

Leukemia

in vitro

K-562

[118]

Irinotecan

Cancers 13 05346 i083

Liposome

Colon

in vivo

Colo 320DM and Colon 26 xenograft mice

[119]

SLN

Rectal, Colon

in vivo

SCC7 xenograft mice

[120]

Isoquinoline

Berberine

Cancers 13 05346 i084

Liposome

Hepatic

in vitro; in vivo

HepG2/HepG2 xenograft mice

[121]

Breast

in vitro; in vivo

MCF-7/MCF-7 CSC xenograft mice

[122]

SLN

Breast, Hepatic, Lung

in vitro

MCF-7, HepG2, A549

[123]

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021.
  2. Bryant, A.K.; Banegas, M.P.; Martinez, M.E.; Mell, L.K.; Murphy, J.D. Trends in Radiation Therapy among Cancer Survivors in the United States, 2000–2030. Cancer Epidemiol. Biomark. Prev. 2017, 26, 963–970.
  3. Perera, S.K.; Jacob, S.; Sullivan, R.; Barton, M. Evidence-based benchmarks for use of cancer surgery in high-income countries: A population-based analysis. Lancet Oncol. 2021, 22, 173–181.
  4. Wilson, B.E.; Jacob, S.; Yap, M.L.; Ferlay, J.; Bray, F.; Barton, M.B. Estimates of global chemotherapy demands and corresponding physician workforce requirements for 2018 and 2040: A population-based study. Lancet Oncol. 2019, 20, 769–780.
  5. Falzone, L.; Salomone, S.; Libra, M. Evolution of Cancer Pharmacological Treatments at the Turn of the Third Millennium. Front. Pharmacol. 2018, 9, 1300.
  6. Sharma, P.; Hu-Lieskovan, S.; Wargo, J.A.; Ribas, A. Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell 2017, 168, 707–723.
  7. Li, F.S.; Weng, J.K. Demystifying traditional herbal medicine with modern approach. Nat. Plants 2017, 3, 17109.
  8. Atanasov, A.G.; Zotchev, S.B.; Dirsch, V.M.; International Natural Product Sciences Taskforce; Supuran, C.T. Natural products in drug discovery: Advances and opportunities. Nat. Rev. Drug Discov. 2021, 20, 200–216.
  9. Newman, D.J.; Cragg, G.M. Natural Products as Sources of New Drugs from 1981 to 2014. J. Nat. Prod. 2016, 79, 629–661.
  10. Davison, E.K.; Brimble, M.A. Natural product derived privileged scaffolds in drug discovery. Curr. Opin Chem Biol. 2019, 52, 1–8.
  11. Eguchi, R.; Ono, N.; Hirai Morita, A.; Katsuragi, T.; Nakamura, S.; Huang, M.; Altaf-Ul-Amin, M.; Kanaya, S. Classification of alkaloids according to the starting substances of their biosynthetic pathways using graph convolutional neural networks. BMC Bioinform. 2019, 20, 380.
  12. Ehrenworth, A.M.; Peralta-Yahya, P. Accelerating the semisynthesis of alkaloid-based drugs through metabolic engineering. Nat. Chem. Biol. 2017, 13, 249–258.
  13. Bharate, S.S.; Mignani, S.; Vishwakarma, R.A. Why Are the Majority of Active Compounds in the CNS Domain Natural Products? A Critical Analysis. J. Med. Chem. 2018, 61, 10345–10374.
  14. Reuvers, T.G.A.; Kanaar, R.; Nonnekens, J. DNA Damage-Inducing Anticancer Therapies: From Global to Precision Damage. Cancers 2020, 12, 2098.
  15. Steinmetz, M.O.; Prota, A.E. Microtubule-Targeting Agents: Strategies to Hijack the Cytoskeleton. Trends Cell Biol. 2018, 28, 776–792.
  16. Douer, D. Efficacy and Safety of Vincristine Sulfate Liposome Injection in the Treatment of Adult Acute Lymphocytic Leukemia. Oncologist 2016, 21, 840–847.
  17. Innocenti, F.; Schilsky, R.L.; Ramirez, J.; Janisch, L.; Undevia, S.; House, L.K.; Das, S.; Wu, K.; Turcich, M.; Marsh, R.; et al. Dose-finding and pharmacokinetic study to optimize the dosing of irinotecan according to the UGT1A1 genotype of patients with cancer. J. Clin. Oncol. 2014, 32, 2328–2334.
  18. Rosello, S.; Blasco, I.; Garcia Fabregat, L.; Cervantes, A.; Jordan, K.; Committee, E.G. Management of infusion reactions to systemic anticancer therapy: ESMO Clinical Practice Guidelines. Ann. Oncol. 2017, 28, iv100–iv118.
  19. Blanco, E.; Shen, H.; Ferrari, M. Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat. Biotechnol. 2015, 33, 941–951.
  20. Pelaz, B.; Alexiou, C.; Alvarez-Puebla, R.A.; Alves, F.; Andrews, A.M.; Ashraf, S.; Balogh, L.P.; Ballerini, L.; Bestetti, A.; Brendel, C.; et al. Diverse Applications of Nanomedicine. ACS Nano 2017, 11, 2313–2381.
  21. Tang, W.; Fan, W.; Lau, J.; Deng, L.; Shen, Z.; Chen, X. Emerging blood-brain-barrier-crossing nanotechnology for brain cancer theranostics. Chem. Soc. Rev. 2019, 48, 2967–3014.
  22. Lancet, J.E.; Uy, G.L.; Cortes, J.E.; Newell, L.F.; Lin, T.L.; Ritchie, E.K.; Stuart, R.K.; Strickland, S.A.; Hogge, D.; Solomon, S.R.; et al. CPX-351 (cytarabine and daunorubicin) Liposome for Injection Versus Conventional Cytarabine Plus Daunorubicin in Older Patients With Newly Diagnosed Secondary Acute Myeloid Leukemia. J. Clin. Oncol. 2018, 36, 2684–2692.
  23. Pattni, B.S.; Chupin, V.V.; Torchilin, V.P. New Developments in Liposomal Drug Delivery. Chem. Rev. 2015, 115, 10938–10966.
  24. Suk, J.S.; Xu, Q.; Kim, N.; Hanes, J.; Ensign, L.M. PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv. Drug Deliv. Rev. 2016, 99, 28–51.
  25. Shi, J.; Kantoff, P.W.; Wooster, R.; Farokhzad, O.C. Cancer nanomedicine: Progress, challenges and opportunities. Nat. Rev. Cancer 2017, 17, 20–37.
  26. Mitchell, M.J.; Billingsley, M.M.; Haley, R.M.; Wechsler, M.E.; Peppas, N.A.; Langer, R. Engineering precision nanoparticles for drug delivery. Nat. Rev. Drug Discov. 2021, 20, 101–124.
  27. Bayda, S.; Adeel, M.; Tuccinardi, T.; Cordani, M.; Rizzolio, F. The History of Nanoscience and Nanotechnology: From Chemical-Physical Applications to Nanomedicine. Molecules 2019, 25, 112.
  28. Roco, M.C. The long view of nanotechnology development: The National Nanotechnology Initiative at 10 years. J. Nanopart. Res. 2011, 13, 427–445.
  29. Porter, A.L.; Youtie, J. How interdisciplinary is nanotechnology? J. Nanopart. Res. 2009, 11, 1023–1041.
  30. Chen, H.; Gu, Z.; An, H.; Chen, C.; Chen, J.; Cui, R.; Chen, S.; Chen, W.; Chen, X.; Chen, X.; et al. Precise nanomedicine for intelligent therapy of cancer. Sci. China Chem. 2018, 61, 1503–1552.
  31. Van der Meel, R.; Sulheim, E.; Shi, Y.; Kiessling, F.; Mulder, W.J.M.; Lammers, T. Smart cancer nanomedicine. Nat. Nanotechnol. 2019, 14, 1007–1017.
  32. Zaytseva, O.; Neumann, G. Carbon nanomaterials: Production, impact on plant development, agricultural and environmental applications. Chem. Biol. Technol. Agric. 2016, 3, 17.
  33. Liu, Z.; Robinson, J.T.; Tabakman, S.M.; Yang, K.; Dai, H. Carbon materials for drug delivery & cancer therapy. Mater. Today 2011, 14, 316–323.
  34. Patel, K.D.; Singh, R.K.; Kim, H.-W. Carbon-based nanomaterials as an emerging platform for theranostics. Mater. Horiz. 2019, 6, 434–469.
  35. Mody, V.V.; Siwale, R.; Singh, A.; Mody, H.R. Introduction to metallic nanoparticles. J. Pharm. Bioallied. Sci. 2010, 2, 282–289.
  36. Saleh, T.A. Nanomaterials: Classification, properties, and environmental toxicities. Environ. Technol. Innov. 2020, 20, 101067.
  37. Palazzolo, S.; Bayda, S.; Hadla, M.; Caligiuri, I.; Corona, G.; Toffoli, G.; Rizzolio, F. The Clinical Translation of Organic Nanomaterials for Cancer Therapy: A Focus on Polymeric Nanoparticles, Micelles, Liposomes and Exosomes. Curr. Med. Chem. 2018, 25, 4224–4268.
  38. Yang, L.; Stockigt, J. Trends for diverse production strategies of plant medicinal alkaloids. Nat. Prod. Rep. 2010, 27, 1469–1479.
  39. Conroy, T.; Hammel, P.; Hebbar, M.; Ben Abdelghani, M.; Wei, A.C.; Raoul, J.L.; Chone, L.; Francois, E.; Artru, P.; Biagi, J.J.; et al. FOLFIRINOX or Gemcitabine as Adjuvant Therapy for Pancreatic Cancer. N. Engl. J. Med. 2018, 379, 2395–2406.
  40. Socinski, M.A.; Jotte, R.M.; Cappuzzo, F.; Orlandi, F.; Stroyakovskiy, D.; Nogami, N.; Rodriguez-Abreu, D.; Moro-Sibilot, D.; Thomas, C.A.; Barlesi, F.; et al. Atezolizumab for First-Line Treatment of Metastatic Nonsquamous NSCLC. N. Engl. J. Med. 2018, 378, 2288–2301.
  41. Cortinovis, D.; Bidoli, P.; Canova, S.; Colonese, F.; Gemelli, M.; Lavitrano, M.L.; Banna, G.L.; Liu, S.V.; Morabito, A. Novel Cytotoxic Chemotherapies in Small Cell Lung Carcinoma. Cancers 2021, 13, 1152.
  42. Sweeney, C.J.; Chen, Y.H.; Carducci, M.; Liu, G.; Jarrard, D.F.; Eisenberger, M.; Wong, Y.N.; Hahn, N.; Kohli, M.; Cooney, M.M.; et al. Chemohormonal Therapy in Metastatic Hormone-Sensitive Prostate Cancer. N. Engl. J. Med. 2015, 373, 737–746.
  43. Connors, J.M.; Jurczak, W.; Straus, D.J.; Ansell, S.M.; Kim, W.S.; Gallamini, A.; Younes, A.; Alekseev, S.; Illes, A.; Picardi, M.; et al. Brentuximab Vedotin with Chemotherapy for Stage III or IV Hodgkin’s Lymphoma. N. Engl. J. Med. 2018, 378, 331–344.
  44. Silva, C.O.; Pinho, J.O.; Lopes, J.M.; Almeida, A.J.; Gaspar, M.M.; Reis, C. Current Trends in Cancer Nanotheranostics: Metallic, Polymeric, and Lipid-Based Systems. Pharmaceutics 2019, 11, 22.
  45. Lim, S.B.; Banerjee, A.; Önyüksel, H. Improvement of drug safety by the use of lipid-based nanocarriers. J. Control Release 2012, 163, 34–45.
  46. Rahman, M.A.; Hussain, A.; Hussain, M.S.; Mirza, M.A.; Iqbal, Z. Role of excipients in successful development of self-emulsifying/microemulsifying drug delivery system (SEDDS/SMEDDS). Drug Dev. Ind. Pharm. 2013, 39, 1–19.
  47. Zhigaltsev, I.V.; Tam, Y.K.; Leung, A.K.K.; Cullis, P.R. Production of limit size nanoliposomal systems with potential utility as ultra-small drug delivery agents. J. Liposome Res. 2016, 26, 96–102.
  48. Cabral, H.; Matsumoto, Y.; Mizuno, K.; Chen, Q.; Murakami, M.; Kimura, M.; Terada, Y.; Kano, M.R.; Miyazono, K.; Uesaka, M.; et al. Accumulation of sub-100 nm polymeric micelles in poorly permeable tumours depends on size. Nat. Nanotechnol. 2011, 6, 815–823.
  49. Silverman, J.A.; Deitcher, S.R. Marqibo(R) (vincristine sulfate liposome injection) improves the pharmacokinetics and pharmacodynamics of vincristine. Cancer Chemother. Pharm. 2013, 71, 555–564.
  50. Barenholz, Y. Doxil®—The first FDA-approved nano-drug: Lessons learned. J. Control Release 2012, 160, 117–134.
  51. Green, M.R.; Manikhas, G.M.; Orlov, S.; Afanasyev, B.; Makhson, A.M.; Bhar, P.; Hawkins, M.J. Abraxane, a novel Cremophor-free, albumin-bound particle form of paclitaxel for the treatment of advanced non-small-cell lung cancer. Ann. Oncol. 2006, 17, 1263–1268.
  52. Sercombe, L.; Veerati, T.; Moheimani, F.; Wu, S.Y.; Sood, A.K.; Hua, S. Advances and Challenges of Liposome Assisted Drug Delivery. Front. Pharm. 2015, 6, 286.
  53. Northfelt, D.W.; Dezube, B.J.; Thommes, J.A.; Miller, B.J.; Fischl, M.A.; Friedman-Kien, A.; Kaplan, L.D.; Du Mond, C.; Mamelok, R.D.; Henry, D.H. Pegylated-liposomal doxorubicin versus doxorubicin, bleomycin, and vincristine in the treatment of AIDS-related Kaposi’s sarcoma: Results of a randomized phase III clinical trial. J. Clin. Oncol. 1998, 16, 2445–2451.
  54. Yingchoncharoen, P.; Kalinowski, D.S.; Richardson, D.R. Lipid-Based Drug Delivery Systems in Cancer Therapy: What Is Available and What Is Yet to Come. Pharm. Rev. 2016, 68, 701–787.
  55. Amoabediny, G.; Haghiralsadat, F.; Naderinezhad, S.; Helder, M.N.; Akhoundi Kharanaghi, E.; Mohammadnejad Arough, J.; Zandieh-Doulabi, B. Overview of preparation methods of polymeric and lipid-based (niosome, solid lipid, liposome) nanoparticles: A comprehensive review. Int. J. Polym. Mater. Polym. Biomater. 2018, 67, 383–400.
  56. Gabizon, A.; Shmeeda, H.; Grenader, T. Pharmacological basis of pegylated liposomal doxorubicin: Impact on cancer therapy. Eur. J. Pharm. Sci. 2012, 45, 388–398.
  57. Steffes, V.M.; Zhang, Z.; MacDonald, S.; Crowe, J.; Ewert, K.K.; Carragher, B.; Potter, C.S.; Safinya, C.R. PEGylation of Paclitaxel-Loaded Cationic Liposomes Drives Steric Stabilization of Bicelles and Vesicles thereby Enhancing Delivery and Cytotoxicity to Human Cancer Cells. ACS Appl. Mater. Interfaces 2020, 12, 151–162.
  58. Lin, W.; Kampf, N.; Goldberg, R.; Driver, M.J.; Klein, J. Poly-phosphocholinated Liposomes Form Stable Superlubrication Vectors. Langmuir 2019, 35, 6048–6054.
  59. Cao, Z.; Zhang, L.; Jiang, S. Superhydrophilic Zwitterionic Polymers Stabilize Liposomes. Langmuir 2012, 28, 11625–11632.
  60. Perez-Herrero, E.; Fernandez-Medarde, A. Advanced targeted therapies in cancer: Drug nanocarriers, the future of chemotherapy. Eur. J. Pharm. Biopharm. 2015, 93, 52–79.
  61. Lewrick, F.; Süss, R. Remote loading of anthracyclines into liposomes. Methods Mol. Biol. 2010, 605, 139–145.
  62. Hardiansyah, A.; Huang, L.Y.; Yang, M.C.; Liu, T.Y.; Tsai, S.C.; Yang, C.Y.; Kuo, C.Y.; Chan, T.Y.; Zou, H.M.; Lian, W.N.; et al. Magnetic liposomes for colorectal cancer cells therapy by high-frequency magnetic field treatment. Nanoscale Res. Lett. 2014, 9, 497.
  63. Park, J.H.; Cho, H.J.; Yoon, H.Y.; Yoon, I.S.; Ko, S.H.; Shim, J.S.; Cho, J.H.; Park, J.H.; Kim, K.; Kwon, I.C.; et al. Hyaluronic acid derivative-coated nanohybrid liposomes for cancer imaging and drug delivery. J. Control Release 2014, 174, 98–108.
  64. Deshpande, P.; Jhaveri, A.; Pattni, B.; Biswas, S.; Torchilin, V. Transferrin and octaarginine modified dual-functional liposomes with improved cancer cell targeting and enhanced intracellular delivery for the treatment of ovarian cancer. Drug Deliv. 2018, 25, 517–532.
  65. Ninomiya, K.; Kawabata, S.; Tashita, H.; Shimizu, N. Ultrasound-mediated drug delivery using liposomes modified with a thermosensitive polymer. Ultrason. Sonochem. 2014, 21, 310–316.
  66. Sesarman, A.; Tefas, L.; Sylvester, B.; Licarete, E.; Rauca, V.; Luput, L.; Patras, L.; Porav, S.; Banciu, M.; Porfire, A. Co-delivery of curcumin and doxorubicin in PEGylated liposomes favored the antineoplastic C26 murine colon carcinoma microenvironment. Drug Deliv. Transl. Res. 2019, 9, 260–272.
  67. Legut, M.; Lipka, D.; Filipczak, N.; Piwoni, A.; Kozubek, A.; Gubernator, J. Anacardic acid enhances the anticancer activity of liposomal mitoxantrone towards melanoma cell lines - in vitro studies. Int. J. Nanomed. 2014, 9, 653–668.
  68. Lakkadwala, S.; Singh, J. Dual Functionalized 5-Fluorouracil Liposomes as Highly Efficient Nanomedicine for Glioblastoma Treatment as Assessed in an In Vitro Brain Tumor Model. J. Pharm. Sci. 2018, 107, 2902–2913.
  69. Ge, X.; Wei, M.; He, S.; Yuan, W.-E. Advances of Non-Ionic Surfactant Vesicles (Niosomes) and Their Application in Drug Delivery. Pharmaceutics 2019, 11, 55.
  70. Ag Seleci, D.; Seleci, M.; Walter, J.-G.; Stahl, F.; Scheper, T. Niosomes as Nanoparticular Drug Carriers: Fundamentals and Recent Applications. J. Nanomater. 2016, 2016, 7372306.
  71. Gharbavi, M.; Amani, J.; Kheiri-Manjili, H.; Danafar, H.; Sharafi, A. Niosome: A Promising Nanocarrier for Natural Drug Delivery through Blood-Brain Barrier. Adv. Pharmacol. Sci. 2018, 2018, 6847971.
  72. You, L.; Liu, X.; Fang, Z.; Xu, Q.; Zhang, Q. Synthesis of multifunctional nano-niosomes as a targeting carrier for treatment of cervical cancer. Mater. Sci. Eng. C Mater. Biol. Appl. 2019, 94, 291–302.
  73. Abdulbaqi, I.M.; Darwis, Y.; Khan, N.A.; Assi, R.A.; Khan, A.A. Ethosomal nanocarriers: The impact of constituents and formulation techniques on ethosomal properties, in vivo studies, and clinical trials. Int. J. Nanomed. 2016, 11, 2279–2304.
  74. Nainwal, N.; Jawla, S.; Singh, R.; Saharan, V.A. Transdermal applications of ethosomes—A detailed review. J. Liposome Res. 2019, 29, 103–113.
  75. Chen, M.; Liu, X.; Fahr, A. Skin penetration and deposition of carboxyfluorescein and temoporfin from different lipid vesicular systems: In vitro study with finite and infinite dosage application. Int. J. Pharm. 2011, 408, 223–234.
  76. Yu, X.; Du, L.; Li, Y.; Fu, G.; Jin, Y. Improved anti-melanoma effect of a transdermal mitoxantrone ethosome gel. Biomed. Pharm. 2015, 73, 6–11.
  77. Mishra, D.K.; Shandilya, R.; Mishra, P.K. Lipid based nanocarriers: A translational perspective. Nanomedicine 2018, 14, 2023–2050.
  78. Lu, Y.; Zhang, E.; Yang, J.; Cao, Z. Strategies to improve micelle stability for drug delivery. Nano Res. 2018, 11, 4985–4998.
  79. Koo, O.M.Y.; Rubinstein, I.; Onyüksel, H. Actively targeted low-dose camptothecin as a safe, long-acting, disease-modifying nanomedicine for rheumatoid arthritis. Pharm. Res. 2011, 28, 776–787.
  80. Krishnadas, A.; Rubinstein, I.; Önyüksel, H. Sterically Stabilized Phospholipid Mixed Micelles: In Vitro Evaluation as a Novel Carrier for Water-Insoluble Drugs. Pharm. Res. 2003, 20, 297–302.
  81. Tang, N.; Du, G.; Wang, N.; Liu, C.; Hang, H.; Liang, W. Improving penetration in tumors with nanoassemblies of phospholipids and doxorubicin. J. Natl. Cancer Inst. 2007, 99, 1004–1015.
  82. Kim, S.; Shi, Y.; Kim, J.Y.; Park, K.; Cheng, J.X. Overcoming the barriers in micellar drug delivery: Loading efficiency, in vivo stability, and micelle-cell interaction. Expert Opin. Drug Deliv. 2010, 7, 49–62.
  83. Alberts, B.; Johnson, A.; Lewis, J.; Raff, M.; Roberts, K.; Peter, W. The Lipid Bilayer. In Molecular Biology of the Cell, 4th ed.; Garland Science: New York, NY, USA, 2002.
  84. Muller, R.H.; Shegokar, R.; Keck, C.M. 20 years of lipid nanoparticles (SLN and NLC): Present state of development and industrial applications. Curr. Drug Discov. Technol. 2011, 8, 207–227.
  85. Uner, M.; Yener, G. Importance of solid lipid nanoparticles (SLN) in various administration routes and future perspectives. Int. J. Nanomed. 2007, 2, 289–300.
  86. Wang, W.; Zhang, L.; Chen, T.; Guo, W.; Bao, X.; Wang, D.; Ren, B.; Wang, H.; Li, Y.; Wang, Y.; et al. Anticancer Effects of Resveratrol-Loaded Solid Lipid Nanoparticles on Human Breast Cancer Cells. Molecules 2017, 22, 1814.
  87. Ghasemiyeh, P.; Mohammadi-Samani, S. Solid lipid nanoparticles and nanostructured lipid carriers as novel drug delivery systems: Applications, advantages and disadvantages. Res. Pharm. Sci. 2018, 13, 288–303.
  88. Muller, R.H.; Radtke, M.; Wissing, S.A. Nanostructured lipid matrices for improved microencapsulation of drugs. Int. J. Pharm. 2002, 242, 121–128.
  89. Beloqui, A.; Solinis, M.A.; Rodriguez-Gascon, A.; Almeida, A.J.; Preat, V. Nanostructured lipid carriers: Promising drug delivery systems for future clinics. Nanomedicine 2016, 12, 143–161.
  90. Iqbal, M.A.; Md, S.; Sahni, J.K.; Baboota, S.; Dang, S.; Ali, J. Nanostructured lipid carriers system: Recent advances in drug delivery. J. Drug Target 2012, 20, 813–830.
  91. Wei, Q.; Yang, Q.; Wang, Q.; Sun, C.; Zhu, Y.; Niu, Y.; Yu, J.; Xu, X. Formulation, Characterization, and Pharmacokinetic Studies of 6-Gingerol-Loaded Nanostructured Lipid Carriers. AAPS PharmSciTech 2018, 19, 3661–3669.
  92. Vakili-Ghartavol, R.; Rezayat, S.M.; Faridi-Majidi, R.; Sadri, K.; Jaafari, M.R. Optimization of Docetaxel Loading Conditions in Liposomes: Proposing potential products for metastatic breast carcinoma chemotherapy. Sci. Rep. 2020, 10, 5569.
  93. Zhigaltsev, I.V.; Winters, G.; Srinivasulu, M.; Crawford, J.; Wong, M.; Amankwa, L.; Waterhouse, D.; Masin, D.; Webb, M.; Harasym, N.; et al. Development of a weak-base docetaxel derivative that can be loaded into lipid nanoparticles. J. Control Release 2010, 144, 332–340.
  94. Chang, M.; Lu, S.; Zhang, F.; Zuo, T.; Guan, Y.; Wei, T.; Shao, W.; Lin, G. RGD-modified pH-sensitive liposomes for docetaxel tumor targeting. Colloids Surf. B Biointerfaces 2015, 129, 175–182.
  95. Ren, G.; Liu, D.; Guo, W.; Wang, M.; Wu, C.; Guo, M.; Ai, X.; Wang, Y.; He, Z. Docetaxel prodrug liposomes for tumor therapy: Characterization, in vitro and in vivo evaluation. Drug Deliv. 2016, 23, 1272–1281.
  96. Luo, Q.; Yang, B.; Tao, W.; Li, J.; Kou, L.; Lian, H.; Che, X.; He, Z.; Sun, J. ATB(0,+) transporter-mediated targeting delivery to human lung cancer cells via aspartate-modified docetaxel-loading stealth liposomes. Biomater. Sci. 2017, 5, 295–304.
  97. Gu, Z.; Wang, Q.; Shi, Y.; Huang, Y.; Zhang, J.; Zhang, X.; Lin, G. Nanotechnology-mediated immunochemotherapy combined with docetaxel and PD-L1 antibody increase therapeutic effects and decrease systemic toxicity. J. Control Release 2018, 286, 369–380.
  98. Mi, Y.; Liu, Y.; Feng, S.S. Formulation of Docetaxel by folic acid-conjugated d-alpha-tocopheryl polyethylene glycol succinate 2000 (Vitamin E TPGS(2k)) micelles for targeted and synergistic chemotherapy. Biomaterials 2011, 32, 4058–4066.
  99. Shi, C.; Zhang, Z.; Wang, F.; Ji, X.; Zhao, Z.; Luan, Y. Docetaxel-loaded PEO-PPO-PCL/TPGS mixed micelles for overcoming multidrug resistance and enhancing antitumor efficacy. J. Mater. Chem. B 2015, 3, 4259–4271.
  100. Liu, H.; Tu, L.; Zhou, Y.; Dang, Z.; Wang, L.; Du, J.; Feng, J.; Hu, K. Improved Bioavailability and Antitumor Effect of Docetaxel by TPGS Modified Proniosomes: In Vitro and In Vivo Evaluations. Sci. Rep. 2017, 7, 43372.
  101. Liu, D.; Liu, Z.; Wang, L.; Zhang, C.; Zhang, N. Nanostructured lipid carriers as novel carrier for parenteral delivery of docetaxel. Colloids Surf. B Biointerfaces 2011, 85, 262–269.
  102. Ravar, F.; Saadat, E.; Gholami, M.; Dehghankelishadi, P.; Mahdavi, M.; Azami, S.; Dorkoosh, F.A. Hyaluronic acid-coated liposomes for targeted delivery of paclitaxel, in-vitro characterization and in-vivo evaluation. J. Control Release 2016, 229, 10–22.
  103. Wang, J.; Jia, J.; Liu, J.; He, H.; Zhang, W.; Li, Z. Tumor targeting effects of a novel modified paclitaxel-loaded discoidal mimic high density lipoproteins. Drug Deliv. 2013, 20, 356–363.
  104. Miao, Y.Q.; Chen, M.S.; Zhou, X.; Guo, L.M.; Zhu, J.J.; Wang, R.; Zhang, X.X.; Gan, Y. Chitosan oligosaccharide modified liposomes enhance lung cancer delivery of paclitaxel. Acta Pharm. Sin. 2021.
  105. Jain, S.; Kumar, D.; Swarnakar, N.K.; Thanki, K. Polyelectrolyte stabilized multilayered liposomes for oral delivery of paclitaxel. Biomaterials 2012, 33, 6758–6768.
  106. Zhang, X.G.; Miao, J.; Dai, Y.Q.; Du, Y.Z.; Yuan, H.; Hu, F.Q. Reversal activity of nanostructured lipid carriers loading cytotoxic drug in multi-drug resistant cancer cells. Int. J. Pharm. 2008, 361, 239–244.
  107. Paolino, D.; Celia, C.; Trapasso, E.; Cilurzo, F.; Fresta, M. Paclitaxel-loaded ethosomes(R): Potential treatment of squamous cell carcinoma, a malignant transformation of actinic keratoses. Eur. J. Pharm. Biopharm. 2012, 81, 102–112.
  108. Zhao, B.J.; Ke, X.Y.; Huang, Y.; Chen, X.M.; Zhao, X.; Zhao, B.X.; Lu, W.L.; Lou, J.N.; Zhang, X.; Zhang, Q. The antiangiogenic efficacy of NGR-modified PEG-DSPE micelles containing paclitaxel (NGR-M-PTX) for the treatment of glioma in rats. J. Drug Target. 2011, 19, 382–390.
  109. Song, X.L.; Liu, S.; Jiang, Y.; Gu, L.Y.; Xiao, Y.; Wang, X.; Cheng, L.; Li, X.T. Targeting vincristine plus tetrandrine liposomes modified with DSPE-PEG2000-transferrin in treatment of brain glioma. Eur. J. Pharm. Sci. 2017, 96, 129–140.
  110. Wang, C.; Feng, L.; Yang, X.; Wang, F.; Lu, W. Folic acid-conjugated liposomal vincristine for multidrug resistant cancer therapy. Asian J. Pharm. Sci. 2013, 8, 118–127.
  111. Aboutaleb, E.; Atyabi, F.; Khoshayand, M.R.; Vatanara, A.R.; Ostad, S.N.; Kobarfard, F.; Dinarvand, R. Improved brain delivery of vincristine using dextran sulfate complex solid lipid nanoparticles: Optimization and in vivo evaluation. J. Biomed. Mater. Res. A 2014, 102, 2125–2136.
  112. Gao, X.; Zhang, J.; Xu, Q.; Huang, Z.; Wang, Y.; Shen, Q. Hyaluronic acid-coated cationic nanostructured lipid carriers for oral vincristine sulfate delivery. Drug Dev. Ind. Pharm. 2017, 43, 661–667.
  113. Li, X.T.; He, M.L.; Zhou, Z.Y.; Jiang, Y.; Cheng, L. The antitumor activity of PNA modified vinblastine cationic liposomes on Lewis lung tumor cells: In vitro and in vivo evaluation. Int. J. Pharm. 2015, 487, 223–233.
  114. Amiri, B.; Ahmadvand, H.; Farhadi, A.; Najmafshar, A.; Chiani, M.; Norouzian, D. Delivery of vinblastine-containing niosomes results in potent in vitro/in vivo cytotoxicity on tumor cells. Drug Dev. Ind. Pharm. 2018, 44, 1371–1376.
  115. Bahadori, F.; Topcu, G.; Eroglu, M.S.; Onyuksel, H. A new lipid-based nano formulation of vinorelbine. AAPS Pharm. Sci. Tech. 2014, 15, 1138–1148.
  116. Dadashzadeh, S.; Vali, A.M.; Rezaie, M. The effect of PEG coating on in vitro cytotoxicity and in vivo disposition of topotecan loaded liposomes in rats. Int. J. Pharm. 2008, 353, 251–259.
  117. Yu, Y.; Wang, Z.H.; Zhang, L.; Yao, H.J.; Zhang, Y.; Li, R.J.; Ju, R.J.; Wang, X.X.; Zhou, J.; Li, N.; et al. Mitochondrial targeting topotecan-loaded liposomes for treating drug-resistant breast cancer and inhibiting invasive metastases of melanoma. Biomaterials 2012, 33, 1808–1820.
  118. Souza, L.G.; Silva, E.J.; Martins, A.L.; Mota, M.F.; Braga, R.C.; Lima, E.M.; Valadares, M.C.; Taveira, S.F.; Marreto, R.N. Development of topotecan loaded lipid nanoparticles for chemical stabilization and prolonged release. Eur. J. Pharm. Biopharm. 2011, 79, 189–196.
  119. Hattori, Y.; Shi, L.; Ding, W.; Koga, K.; Kawano, K.; Hakoshima, M.; Maitani, Y. Novel irinotecan-loaded liposome using phytic acid with high therapeutic efficacy for colon tumors. J. Control Release 2009, 136, 30–37.
  120. Din, F.U.; Choi, J.Y.; Kim, D.W.; Mustapha, O.; Kim, D.S.; Thapa, R.K.; Ku, S.K.; Youn, Y.S.; Oh, K.T.; Yong, C.S.; et al. Irinotecan-encapsulated double-reverse thermosensitive nanocarrier system for rectal administration. Drug Deliv. 2017, 24, 502–510.
  121. Lin, Y.C.; Kuo, J.Y.; Hsu, C.C.; Tsai, W.C.; Li, W.C.; Yu, M.C.; Wen, H.W. Optimizing manufacture of liposomal berberine with evaluation of its antihepatoma effects in a murine xenograft model. Int. J. Pharm. 2013, 441, 381–388.
  122. Ma, X.; Zhou, J.; Zhang, C.X.; Li, X.Y.; Li, N.; Ju, R.J.; Shi, J.F.; Sun, M.G.; Zhao, W.Y.; Mu, L.M.; et al. Modulation of drug-resistant membrane and apoptosis proteins of breast cancer stem cells by targeting berberine liposomes. Biomaterials 2013, 34, 4452–4465.
  123. Wang, L.; Li, H.; Wang, S.; Liu, R.; Wu, Z.; Wang, C.; Wang, Y.; Chen, M. Enhancing the antitumor activity of berberine hydrochloride by solid lipid nanoparticle encapsulation. AAPS PharmSciTech 2014, 15, 834–844.
More
Information
Subjects: Oncology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 1.3K
Revisions: 2 times (View History)
Update Date: 11 Nov 2021
1000/1000
Video Production Service