Int J App Pharm, Vol 13, Issue 6, 2021, 1-18Review Article

FUNCTIONALIZED POLYMERIC NANOPARTICLES: A NOVEL TARGETED APPROACH FOR ONCOLOGY CARE

PRASIDDHI R. RAIKAR, PANCHAXARI M. DANDAGI*

Department of Pharmaceutics, KLE College of Pharmacy, Belagavi, A Constituent Unit of KLE Academy of Higher Education and Research, Belagavi 590010, Karnataka, India
Email: pmdandagi@yahoo.com

Received: 12 Jul 2021, Revised and Accepted: 30 Aug 2021


ABSTRACT

Popular cancer therapies face extreme disadvantages, including multimedicament tolerance and non-target impact. These issues will lead to poorer patient conformity and poor levels of survival. Successful medical therapies for cancer patients are desperately required. Nano-particulate structures with a pluronic base represent revolutionary platforms for anti-cancer agent provision. These structures provide great potential for the advancement of cancer therapy due to their pharmacological properties and sufficient physicochemical characteristics. This review aims to offer a more detailed description of the pluronic drug delivery mechani sms that are currently available and explains pluronic as a medicinal polymer. Hydrophobic payload formulations and updated, targeted distribution mechanisms are explained based on pluronic formulations. This analysis offers a rundown of the current situation art related to the theranostic application of polymer micelles targeting the microenvironment of cancer cells. Some guidelines for the future scope and possible opportunities are also been addressed.

Search criteria: Primary sources such as Medline a principal component of PubMed, an online, searchable, and biomedical and life science research literature database has been used. It brings readers to almost any area of interest with research and journal articles. One of the internet resources of importance to get scientific publications is specialized scientific search engines known as Google Scholar a database of research material that can be searched for. I have used the online electronic access portal of Elsevier, such as Science Direct to its publications. Scopus is the biggest abstract and peer-reviewed literature database for scientific journals, books, and conference work. Keywords like Cancer, Pluronic, Nanoparticles, Chemotherapy, Cancer, Theranostic, Targeted, Micelles, and Core-shell are crucial as they notify search engines of the content of the site.

Range of years: 1992-2020.

Keywords: Cancer, Multimedicament, Tolerance, Nanoparticulate, Pluronic, Drug delivery, Hydrophobic, Micelles, Theranostic, Microenvironment


INTRODUCTION

Every year, cancer causes millions of deaths globally and while there have been significant strides in treatment, several problems need to be solved to strengthen cancer therapy. Therefore, oncological science strives extremely hard to discover alternative and innovative drugs that can mitigate the important side effects that are induced by traditional therapies. Various innovations are being tested or have already been implemented into clinical use in clinical trials. Although nanomedicine helps create biocompatible materials for both diagnostic and therapeutic purposes, the bioengineering of extracellular vesicles and patient cells allowed ad hoc systems and univocal targeting strategies to be designed [1]. Employing nanotechnology in the field of drug delivery has led to the advent of Nano pharmaceuticals. Nano pharmaceuticals are bound to surmount various obstacles that the field of pharmacy is currently facing by offering various advantages thereby, promising potential to formulate advanced medicines with fewer adverse effects [2]. In this study, we will discuss in detail the latest developments in basic and applied nanotechnology, theranostic approach that have led to the development of developed nanoscale materials as ground breaking prototypes for biomedical applications and tailored targeted treatment for cancer [3].

Existing cancer prevention strategies

Cancer is one of the most frequent illnesses globally, killing about seven million a year. For the past two decades, the outlook for cancer therapy has shifted significantly. New developments in the treatment of cancer, which are based largely on tumor molecular characteristics, are emerging. Treatment of cancer has been made more tumor-specific and less toxic by using modern cancer-centric therapy based on medicinal antibodies or small molecules [4]. However, chemotherapy, surgeries, radiation, or a mixture of these drugs are used in the procedures used for cancer. The foregoing are the various advanced cancer treatments.

Chemotherapy

The application of chemotherapy to cure cancer started to decrease the chemical list by improving ways to diagnose it using transplantable tumors in mice at the turn of the 20th century. Since the late 1940s, chemotherapy has been used to cure many different forms of cancer successfully and to increase survival rates [5]. However, it is not very precise in general and thus endangers normal tissue and organs [6]. Nausea, fatigue, hair loss, anemia, diarrhea, constipation, low blood count, fertility, and more are the major harmful effects of chemotherapy. Often chemotherapeutic agents affect the activity of the brain by direct/indirect pathways, but there are assurances for systemic blood-brain barrier therapy of the brain. Chemotherapy has an intensive and chronic effect on cognitive function, but the cause remains unclear [7]. Several medications, such as Gemcitabine, Azacitidine, Pemetrexed, Paclitaxel, Docetaxel, and many others, are used in chemotherapy. Tablets, capsules, and parenteral (intramuscular, intravenous) are the different routes from which anticancer medications are taken [8].

Radiation therapy

The bulk of people who undergo radiation therapy after their course of the disease continue to receive cancer care as a central factor, leading to a 40% cure of cancer. The treatment with radiation takes away the ability of cancer cells to replicate [9]. High-energy radiation in radiation therapy is used to reduce cancers and destroy cancer cells. X-rays, gamma rays, and charged particles are different sources of radiation used for the treatment of cancer. It destroys cancer cells by destroying the molecules known as Deoxyribonucleic Acid (DNA) in cells and conveys genetic information for the killing of cancer cells from generation to generation [10].

Surgical therapy

The turn of the 20th century marked the beginning of the advancement of cancer operating methods, and in 1908 Miles conducted the first abdominoperineal resection and the first lobectomy in 1912 [11]. The current operation improved dramatically, with non-invasive procedures like laparoscopic colectomy (for colon cancer removal) and thoracic video techniques replacing Halstedian procedures [12]. Sentinel node removal has been used to boost esthetic outcomes and to prevent lymphedema [13]. The use of laryngoscopic laser procedures on early laryngeal cancer is another example of traditional surgery [14]. The most recent breakthrough is Da Vinci®, which is a robot device for the treatment of prostate and renal cancer [15].

Proton therapy

It has wonderful promise as a therapy for multiple tumors. Public interest in proton therapy has risen to a large degree since the Food and Drug Administration (FDA) approved it in 2001. In children with multiple cancers, Proton Therapy is most effective in people with organs with tumors such as kidney, bladder, brain, spine, lungs, back, and leg. Proton therapy is more common. Proton Therapy centers continue to assess their use for more cancers in science [16]. Nevertheless, we do not neglect the value of proper patient preparation, precise science analysis, including contrasts with other technologies, ethical challenges, and economic performance [17].

Thermotherapy

Thermotherapy has been used for at least 4000 y to treat tumors and even before that point, to destruct the tumor masses. Extreme temperature (hyperthermia) can cause tumors to break down by destroying tumor cells and damaging proteins and structures within cancer cells [18]. In many clinical trials, hyperthermia has been used in conjunction with radiation therapy and varicose vein treatment chemotherapy. Thermotherapy is a treatment of body tissue with high-temperature penetration up to 113 degrees F [19, 20].

Photodynamic therapy (PDT)

In 1903, the first scientific use of photodynamic therapy in cancer therapies was discovered for eosin to be aimed at basal cell carcinomas (BCC). For photodynamic therapy, a medicament known as a photosensitizing agent or photosensitive agent with a particular wavelength of light is used. Photosensitizing agent emits a kind of oxygen that destroys surrounding cells on exposure to a certain wavelength of light [21, 22]. PDT can be combined with other medications, for example, chemotherapy, radiation, or surgery. Many clinical experiments are ongoing to test the use of PDT for different forms of cancer [23]. The only way to be able to use nanoparticles as a photo-sensitizing carrier is to meet al. l of the needs of an optimal PDT agent [24].

Laser therapy

Lasers are most widely used for cancers and precancerous development to shrink or kill. The most prevalent application with laser therapy is peripheral disorders, including cancer of the skin of basal cells in the very early stages of multiple cancers, such as non-small cell lung cancer, vulvar, ovarian, penile, and cervical cancer. Laser therapy can also mitigate various signs of cancer, such as bleeding or obstruction. In combination with various other therapies, including surgery, chemotherapy, or radiation therapy, laser therapy can be used. Furthermore, laser treatment can be used to scan the lymph vessels to reduce swelling and minimize the metastasis for tumor cells [25]. Three types of laser are used more commonly in different tumor types, including the neodymium of yttrium-aluminum-garnet (Nd: YAG), argon lasers, and carbon dioxide (CO2). Laser therapy may also evaluate nerve endings to relieve pain after treatment [26].

Immunotherapy

Recent advances and clinical tests have shown that adoptive antitumor-infiltering tumor therapy in about 50–75% of Multiple Myeloma (MM) patients will effectively cause tumor regression [27, 28]. Antitumor Tumor-Infiltrating Lymphocyte (TIL) may also be used to expand adoptive cell transplant therapy to treat patients with other types of cancer, including brain, renal, and lung. However, combination therapies such as Dendritic Cell-Cytokine-Induced killer (DC-CIK) in combination therapy in patients with metastatic breast cancer may enhance their longevity free of relapse and overall survival [29]. Tumor regressions in 72 % of patients with metastatic melanoma can result in adoptive cell therapy in tandem with non-myeloablative chemical therapy and complete body irradiation, whereas TIL adoptive immunotherapy with non-myeloablative chemotherapy in only 52 % of treated patients can cause tumor regression [27].

Gene therapy

To regain lost functionality and eliminate viruses, gene therapies seek to cure diseases through the insertion of DNA and Ribonucleic Acid (RNA), minor interfering RNA, and antisense oligonucleotides into special target cells or tissues. The therapeutic genes are supplied to certain target cells with effective vectors aimed at retaining stable, regulated gene expression without causing undesirable side effects [30]. Transforming viruses into genetic shuttles to provide the cell gene of interest is one of the fundamental concepts of gene therapy [31]. Gene transfer is a modern cancer therapy approach that incorporates new genes into a tumor cell or the tissue around to induce apoptosis or delay tumor growth.

Nanotherapeutics

Latest efforts have centered on designing functionalized therapeutic nanoparticles that are over-expressed to various cancer cells for particular molecular purposes. Possible benefits of engineered nanoparticles in terms of therapeutic therapies include the potential to transform undesirable physical and chemical properties of the bioactive molecules into desired biopharmacology patterns; increase therapeutic distribution through biologic boundary areas; monitor bioactive agent release; increase therapeutic effectiveness, through administering therapeutics to biological targets selectively; and, by integrating multimodal imaging and simultaneous testing and treatment, execute theranostic functions on multifunctional platforms. The multifunctional framework focused on pluronic nanoparticles with the potential of integrating imaging with therapy as well as incorporating multiple receptor targeting has been providing new insights using novel nanomaterial’s for cancer care (fig. 1). Moreover, a photothermal approach to the removal of cancer cells or tumor tissue, which may have significant promise in the therapeutic environment, has been included in most research on pluronic nanoparticular cancer therapy [32, 33].

Fig. 1: Biomedical applications of nanotherapeutics

Polymeric nanoparticles

The development of nanoparticle-based clinical treatment methods has resulted in significant pharmacological advancements that have decreased adverse effects and improved the safety, resolvability, Pharmacokinetics, and biodistribution of cytotoxic drugs. Polymeric nanoparticles remain popular in cancer therapy because they are a good platform for the study of hydrophilic as well as hydrophobic drugs [3, 34-35]. But most medicines are released into the extracellular matrix; their efficacy relies on tissue diffusion and their use is limited by their poor in vivo specificity. Therefore, promising progress in cancer science is the latest site-specific targeting of nanoparticles. Table 1 lists several examples of polymeric nanoformulation [36]. To improve the biodistribution of antitumor agents, NPs have been designed for optimal size and surface characteristics to increase their circulation time in the bloodstream. They are able to carry and deliver their active drug payloads to cancer cells, by passive targeting mechanisms, such as the EPR effect as well as by active targeting mechanisms using ligands directed against selected determinants differentially over expressed on the surface of tumor cells [37]. One promising solution is the BIND-014 technology, made from docetaxel loading polymeric nanoparticles able to detect prostate cancer by targeting Prostate-specific membrane antigen (PSMA), prostate cell prostate cancer tumor antigen, and non-prostate solid tumor vasculature. In Phase II clinical trials, BIND-014 has shown greater antitumor efficacy in the lower doses of advanced or metastatic non-small-cell lung cancer than normal documentation for the use of non-small-cell lung cancer [3, 38].

Table 1: Drug-loaded polymer nanoparticles in clinical trials or clinical use

Product Drug Applications Status Reference
Abraxane Paclitaxel Breast cancer, non-small cell lung cancer, pancreatic cancer Approved 36
BA-003 Doxorubicin Hepatocellular carcinoma Phase III
Mitoxantrone-loaded Polybutylcyanacrylate (DHAD-PBCA-NPs) Mitoxantrone Hepatocellular carcinoma Phase II
ProLindac Dichloro(1,2-diaminocyclohexane)platinum(II) DACHPt Advanced ovarian cancer Phase III
ABI-008 Docetaxel Metastatic breast cancer, prostate cancer Phase II
ABI-009 Rapamycin Solid tumors Phase II
ABI-011 Thiocolchicine dimer Solid tumors, lymphoma Phase II
BIND-014 Docetaxel Non-small cell lung cancer Phase II
Cyclosert Camptothecin Solid tumors, rectal cancer, renal cell carcinoma, non-small cell lung cancer Phase II
CALAA-01 siRNA targeting Solid tumors Phase I
Docetaxel-PNP Docetaxel Solid tumors Phase I
Nanotax Paclitaxel Peritoneal neoplasms Phase I

NP targeting strategies

Ideally, the target applies rather than indiscriminate delivery across the entire body, to the precise localization of NPs to the desired site. These target NPs need to resolve external obstacles, path barriers, and cellular barriers before the site being accumulated [39]. There have been two main techniques for targeting tumors–passive and active targeting, shown in fig. 2.

Fig. 2: Schematic representation of passive and active targeting approaches. EPR, enhanced permeability and retention; np, nanoparticle [39]

Passive targeting

It benefits from diseased tissues, usually tumor, pathophysiological properties, while active targeting by the drug carrier initially uses passive targeting to gather in the tumor zone and then bind to the target cells using ligands to internalize the NPs to the cells [40, 41]. The passive targeting of therapeutics from nanocarrier depends on the tumor microenvironment, the enhanced permeability and retention (EPR) effect, and the tumor pH. Tumor cells expand and proliferate more quickly than normal cells are well established. This cell proliferation has a metabolic rate that requires more nutrients and a larger amount of oxygen. The architecture of normal cells is disturbed and replaced by tumor cells to compensate for nutrients [42]. Passive targeting helps NPs to build up in the tissue through EPR [43]. Ground Shift in PEG NPs. Enhanced NP hydrophobicity associated with higher particle-partisan aggregation and blood opsonization steric deficiency was found [44, 45]. Poloxamins, poloxamins, Polyethylene Glycol (PEG), Poly-Caprolactone (PCL), Poly D, L-lactic-co-Glycolic Acid (PLGA) are the polymers widely used in the manufacture of sterically stable stealth nodes and to improve hydrophillicity [46].

Active targeting

The therapeutic medication can be obtained with or without the use of coupling agents by mixing medicine or nanocarrier with a cellular targeting motive known as ligands. These target moieties have a special affinity with cell surface antigens (for example, receptors) and can be differentiated among normal and tumor cells based on levels of receptor or antigen expression [43]. The use of targeted Herceptin NPs helped distinguish positive and negative cell breast cancer epidermal growth factor 2 (HER2) in humans. The successful targeting of HER2 receptors with NPs has been verified in over-expressed cells [47]. In designing a targeted delivery system, the specific properties of cancer cells may be used. For example, cancer cells frequently overexpress tumor antigens, structures similar to carbohydrates, or receptors of the growth factor (e. g., epidermal growth factor receptor). Different ligands may be used as active targeted molecules such as antibodies, polysaccharides, aptamers, peptides, transfers, folate, and other small molecules, based on this definition [48]. Table 2 offers some examples of target ligands connected to NPs and their respective targets. Ligand selection depends on the targeted cells [49].

Table 2: An overview of different targeting ligand decorated PLGA NPs

Nanoparticles type Targeting ligand Loaded drug Cell line/Animal model Reference
PEG-PCL Angiopep-2 Paclitaxel U87 MG, Brain Capillary Endothelial Cells (BCECs) 50
PLGA g7 Peptide Loperamide, Rhodamine-123 Tail vein in rats 51
PLGA Trastuzumab Paclitaxel Caco-2, SK-BR-3 52
PLGA Humanized anti-DC-SIGN (hD1)

Fluorescein Isothiocyanate Titanus Toxoid (FITC-TT) Peptide, Dye Quenched-Bovine Serum Albumin

(DQ-BSA)

Granulocytes, Peripheral Blood Mononuclear Cells (PBMCs) 53
PLGA-PEG A10 Prostate-Specific Membrane Antigen (PSMA) aptamer Cisplatin LNCaP, PC3 54
PLGA-PEG Pep TGN Coumarin-6 bEnd.3 55
PLGA-PEG c-Arginyl Glycylaspartic Acid (RGD) peptide A-10 Doxorubicin

MDA-MB-231,B16F10

MCF-7

56
PLGA-PEG 2-fluoropyrimidine RNA aptamers Docetaxel LNCaP 57
PLGA-PEG Folate binding protein Docetaxel SKOV3 58
PLGA-TPGS Tocopheryl Polyethylene Glycol Succinate (TPGS) Docetaxel Caco-2, MCF-7 59
PLGA-TPGS Vitamin E TPGS-folate Doxorubicin MCF-7, C6 glioma 60

Table 3: Structural features and CMC of some poloxamers (Pluronic®) commercially available

Copolymer Molecular weight (Da) Total average polyethylene oxide units Total average polypropylene oxide units Hydrophilic–lipophilic balance Critical micelle concentration (mm) Reference
L10 3200 7.3 49.7 12–18 --- 71
L35 1900 21.6 16.4 18–23 5.3
F38 4600 83.6 15.9 >24 ---
L42 1630 7.4 22.5 7–12 ---
L43 1850 12.6 22.4 7–12 2.2
L44 2200 20.0 22.8 12–18 3.6
L61 2000 4.55 31.0 1–7 0.11
L62 2500 11.4 34.5 1–7 0.40
L64 2900 26.4 30.0 12–18 0.48
P65 3400 38.6 29.3 12–18 ---
F68 8400 152.7 29.0 >24 0.48
F77 6600 105.0 34.1 >24 ---
L81 2750 6.3 42.7 1–7 0.023
P84 4200 38.2 43.5 12–18 0.071
P85 4600 52.3 39.7 12–18 0.065
F87 7700 122.5 39.8 >24 0.091
F88 11400 207.3 39.3 >24 0.25
L92 3650 16.6 50.3 1–7 0.088
F98 13000 236.4 44.8 >24 0.077
L101 3800 8.6 59.0 1–7 0.0021
P103 4950 33.8 59.7 8–12 0.0061
P104 5900 53.6 61.0 12–18 0.0034
P105 6500 73.9 56.0 12–18 ---
F108 14600 265.5 50.3 >24 0.022
L121 4400 10.0 68.3 1–7 0.0010
L122 5000 22.2 69.0 1–7 ---
P123 5750 39.2 69.4 7–12 0.0044
F127 12,600 200.5 65.2 18–23 0.0028

What are pluronic? 

The Baden Aniline and Soda Factory (BASF), under the trade names Pluronics and Tetronics (also named Poloxamers and polyamines, respectively), commercialized Ethylene oxide-propylene oxides (EO-PO) based block of Copolymers some Decades ago [61]. Poly Ethylene Oxide (PEO) is a hydrophilic section that contributes 70% of the block copolymer, while PEO is a water-soluble nonionic class A-B A and B-A-B triblock copolymers, A is (PEO) and B is polypropylene oxide (PPO), with PPO being hydrophobic, and PPO is a contributing 30% of block copolymer [62]. The monomers of the copolymer blocks (e. g., polar and nonpolar) are chemically distinct and thus the block copolymers are amphiphilic and induce active surface properties. Interesting nanostructures that are spontaneously produced by solution result from the block separation (self-assembly). Based on the PEO water solubility and PPO insolubility, Poloxamers demonstrated an amphiphilic character in aqueous solutions. Thus hydraulic are the PEO bricks, and hydraulic is the PPO stone. In many applications, they have been made useful by their size and composition as well as their adsorption, including drug distribution, nanoparticles synthesis, cosmetics and emulsion formulations, efficient ink/pigment dispersants as flexible anti-biofouling shielding, amongst other items [63-70]. Poloxamers are commonly studied in pharmaceutical trials. Of the numerous pluronic F127 (PF127) types, the wide variety of biomedical applications has provided considerable interest. A full range of molecular weights and PPO/PEO ratios are given with Poloxamers. Examples of commercially available Pluronic® are presented in table 3 [71]. They show strong cell compatibility and do not cause major inflammation following administration (e. g., intraperitoneal) or topical administration [41, 72]. Although PEO-PPO materials do not degrade under conditions of physiology, renal filtration eliminates copolymers with molecular weights lower than 15 kDa [73]. The useful features pave the way for the approval of the United States Food and Drug Administration (US FDA) and European Medicines Evaluation Agency (EMEA) for some linear PEO-PPO-PEO triblock in the food, pharmaceutical, and agricultural industries.

Several drug resistance mechanisms are affected by pluronic

Inhibition of P-glycoprotein (Pgp) drug efflux system

The increased cytotoxicity of Pluronic in anthracycline drug-resistant cancer, doxorubicin, seems related to the effects of copolymers on the transportation of the Pgp drug efflux system. This is confirmed by the observation that the doxorubicin accumulation in intracellular resistant cancer cells which express Pgp can greatly increase [74, 75]. No alterations in the use of drugs in Pluronic presence were identified with non-Pgp-expressing carcinogenic cells, i.e. the Pgp-controlled transportation routes in Multidrug-resistant (MDR) cells were especially affected with copolymers.

Effects on other drug transporters

Proof that pluronic block copolymers alone can be inhibited by the Pgp efflux pump is rising. There is increasing data. In recent years it has been concluded that other organic anion carriers, including Multidrug resistance-associated Protein (MRP2), could also occur in Panc-1, in addition to Pluronic can thus also inhibit these conveyors, resulting in increased aggregation of fluorescein in Panc-1 cells [76].

Effects on drug sequestration within cytoplasmic vesicles

Drug in the MDR cells can be sequestrated inside cytoplasmic vesicles and then expressed from the cell until the drug can function on the cell as expected, which is a further possible challenge in the treatment of tumors that are immune [77-81]. The protection of abnormally high pH gradients across organelle membranes by H1-Adenosine Triphosphate (ATP), a pump based on ATP, achieves drug sequestration in MDR cells [82]. Pluronics were shown to be capable of hyper-Sensitizing multiple MDR tumors, increasing their antitumor activity by 2 to 3 times to the activity of antineoplastic agents. The effect below the CMC was noticed in a dose-dependent analysis and was finally due to the free unimer chains.

Effect on glutathione (GSH) and glutathione S transferases system (GST)

Multidrug resistance-associated protein (MRP) prescription flush transporters are closely related to the detoxification mechanism in MDR cells with respect to several substrates [77]. Studies have also started on GSH/GST device effects of Pluronic block copolymers. For example, after exposure to different pathways for drug resistance, including those cells, to P85, substantial decreases in both GSH and GST intracellular levels were observed in Madin-Darby Canine Kidney (MDCK) cells expressing PRP.

Self-assembly of pluronic

The concentration and temperature of pluronic are mainly modulated in an aqueous medium. In principle and laboratory experiments, the technique has respectively been named lyotropic and thermotropic micellization. If pluronic are combined with water, that's fine for the PEO block and bad for the PPO block; pluronic are micelles in an aqueous shape according to pluronic concentration and the temperature shown in fig. 3 [83]. The pluronic solution behavior with the coarse grain model has been described previously [84, 85]. The L44 monolayer displayed a brush-like behavior where water reached the entire PEO region. Conversely, it seems that the PPO air-region-oriented unit is aimed at reducing water interaction. Of special interest is the fact that all PEO blocks are exposed to water and the central PPO block to the vacuum by means of a U-shape conformation [86]. As the number of unimer exceeds the Critical Micelle Concentration (CMC), micelle, polymer, and lyotropic liquid crystalline phases are formed by self-assembly [87, 88].

The mechanism includes complex molecular exchanges between micelles and bulk solvents and micelles [89]. Very hydrophobic pluronic do not micellize yet at a cloud stage (<20 % PEO) constitute unsteady vesicular structures. The hydrophobic, relatively atomized copolymers form narrowly dissipated micelles of the heart at ambient temperature. Calorimetric differential scanning studies have shown that the migration of unimer to micelles is an endothermic process. PEO block is primarily governed by a temperature-dependent pluronic aggregation. Its character varies between hydrophilic and hydrophobic as the temperature increases, which means critical micelle temperature (CMT) dominates CMC [90]. The hydrophobicity in higher temperatures of the PEO and PPO blocks in combination with the reducing opportunities of H bond formation (the proportion of the anhydrous methyl groups) increases. The shift in the hydrodynamic size of aggregates can be tested for temperature-dependent micellization and de-micellization [91].

Fig. 3: The development of pluronic micelles in the watery medium according to Pluronics and temperature concentration [82]

Modulation in aggregation behavior of pluronic micelles

In the presence of salt, Micelles will typically expand around the cloud point or display a transformation to rod-like assembling [92]. These transitions primarily rely on temperature and additive existence or hydrophobicity and on copolymer concentration. Like electrolytes (inorganic salts, ionic liquids), nonelectrolytes (urea, sugar, alcohol), and amphiphilic compounds (hydrotropes and ionic surfactants with low molecular weight and other copolymers) additives [93-97]. The CMC or CMT is reduced by dehydration of PEO–PPO interface by additives with beneficial interactions with water. On the opposite, micellization of mixed solvents gives greater solvency for copolymers is disadvantaged. For e. g., with the addition of 40v/v percent ethanol in water, Alexandridis and coworkers showed double increases in CMC P105-F127 [98]. Pluronics solution behavior can also be modified by chemical alteration. Depending on demand, neutral hydrophilic blocks can be cationized or anionised [99]. In pentablock copolymers made of poly (N-isopropyl acrylamide) and poly (lactic acid-co-glycolic acid) and separate end blocks, major changes were recorded in step actions [100, 101]. Mixed pluronic structures with ionic surfactants form small micelles that are rich in surfactants and increase surfactant concentration [102]. Mixing of pluronic with other nonionic surfactants containing PEO as hydrophilic block reported synergistic results [103-106].

Drug solubilization in pluronic micelles

Restricted aqueous solubility of medicines remains a problem, especially given the increasing trends in molecular synthesis with high molecular weights, melting points, and lipophilicity [107]. The bioavailability is constrained by the dissolution of poorly soluble anticancer drugs. Through the use of micellar structures, this can be greatly circumvented. Pluronic metabolism is desirable for both topical and systemic administration, owing to its low immunogenic and special core-shell structures. Without embolism risk, micelles may be injected. The hydrophobic center makes it possible to integrate several medicinal products while the hydrophobic corona protects against aggregation, protein adsorption, and soluble locus, and coordination of the loaded molecules depends on their composition and their relative hydrophobicity [108-110]. The first depends primarily on the presence of pluronic, hydrophobic blocks to ensure the superior involvement of drug molecules [111, 112].

Tumor-selective drug targeting with pluronic

The transportation of drug transporters in the extracellular region from tumor interstitial to target cells can be increased with high-affinity interactions. This can be achieved by means of ligands, which display selective binding on cancer cellular surface to an upregulated molecular target. This technique increases cell absorption, off-targeting effects and amplifies clinical benefits by withdrawing the target cells from within. Despite their high specificity, anti-compound targeting is limited in large-scale development due to their large molecular sizes, immunogenic, and complexity. Its large size hinders carrier trafficking, particularly in solid tumors [113]. On the other hand, low molecular weight compounds are inexpensive, non-immunogenic, and have superior regulation of the density on the surface of the carrier. Terminal hydroxyl groups of pluronic were used to attack ligands in literature papers [68, 114-116]. The approach involves the immediate binding of ligands to more volatile aldehyde, carboxylic acids, and primary amine terminals or the derivation of hydroxyl groups. The latter can be used to bind molecules sensitive to stimuli and targets. Crosslinkable groups are often inserted into the hydrophilic block to reduce premature drug-loaded release that may otherwise take place by de-micellizing unnecessarily diluting micelles. Despite phase transition, the pluronic with low CMC values have been selected as modifiers of biological responses, as seen in fig. 4. Pluronics triggered the cytochrome C release with increased cytosolic reactive oxygen levels in the cytoplasm, which resulted in a pro-apoptotic signal being strengthened or a resistance against acupuncture in MDR cells being decreased [117]. Pluronic unimer built into the mitochondrial membranes has also been suggested to alter membrane structure due to elevated cytochrome C and Reactive Oxygen Species (ROS) levels contributing to mitochondrial apoptosis. Pluronic P85 with Doxorubicin (DOX) and Breast Cancer Resistance Protein (BCRP) plays a critical role in increasing the signal pro-apoptotic and restricts the anti-apoptotic cell machinery in cellular levels in MDR cells [118]. However, DOX alone will simultaneously activate both the pro-apoptotic signal and cellular defense against responsive DOX cancer cells [119].

pH-responsive micelles

The carriers have clear pH differences as a result of their path from the blood into the tumor microenvironment, cells, and subset cells. While their environment is a little bit acidic (6.5 to 7.2), pH decreased considerably on endosomes (5.5–6.0) and lysosomes (4.0–5.0) have been reported [120, 121]. The goal is that drug cleavage and release should occur on latter endosomes or lysosomes and tumor tissue [122-124]. Acid-labile bonds including ketal, acetal, hydrazone, imine, cis aconytil, and orthoester were explored. Compared to those focused on the behavior of lysosomal proteases, conjugates constructed with pH-sensitive bonds are superior. The synthetic solution requires the direct attachment of the medication to or trapping in pH-sensitive block micelles using pH. The first was demonstrated by the covalent attachment of curcumin utilizing a cis-aconytil anhydride connector on hydrophilic blocks of F68. The covalent relation of pH-responsive poly (β-amino ester), as a biodegradable polymer, is used in another approach. While low pH makes it possible to release drugs within the cell, the key challenge remains improving micelle affinity for cancer cells. This can be done by the latest demonstration by Xu et al., of identifiable ligands on micelles. A possible target of such transports is tumor cells over-expressing sialic acid residues in the extracellular domain. In contrast with free doxorubicin, hybrid micelles have more than 3.4 times improved the potency of tumor inhibition. On Folic acid attachment, similar changes were observed in the anti-tumor effect of pH-sensitive F127 micelles. Rather recently, as P123 has been connected to a molecule believed to enhance the solubilization of medicine (P115), Tange and coworkers confirm selective oxidative damage to cells of cancer; they have increased intracellular reactive oxygen species, interrupting with mitochondrial activity. Drug molecules escaped the lysosome successfully and were located near the nucleus, which translated into apoptotic death of the tumor cells [125-127].

Fig. 4: Effect of pluronics in MDR cancer cells: (1) entrance of Pluronics, (2) ATP depletion, (3) drug efflux inhibition, (4) cytochrome C release and elevation of ROS in the cytoplasm, (5) pro-apoptotic signaling elevation and lower anti-apoptotic defense, (6) inhibition on the glutathione/glutathione S-transferases detoxification system, and (7) elimination of drug sequestration within cytoplasmic vesicles [116]

Redox sensitive micelles

The uncontrolled proliferation and a high metabolic rate of cancer cells contribute to high reactive oxygen species development. ROS speeds up cancer mutation rates and causes irregular signals [128]. While the intracellular, extracellular redox cells actively configure their state, a safeguard device acts at the same time. The development of GSH, a tripeptide-containing thiol (μ-glutamyl-cysteinyl-glycine), effectively controls the latter. GSH mediates electrical, free radical, and ROS neutralization [129, 130].

Ultrasound-sensitive micelles

The reversible endothelium permeability and focused drug release can be manipulated at the tumor site by acoustic waves. Tumor simulation and drug release activation can be performed concurrently here. The tumor interstitium will discharge a high payload within the pluronic micelles by ultrasound-induced interference. The destruction of micelles and the payload release has been demonstrated that the power output change can be remotely controlled [131]. The copolymer was synthesized using azide-completed PEO and alkyne-completed PPO, which contains 1, 2, 3 triazole movement and four ester bonds at the junction site. The breakage of crossroads caused by High-Intensity Focused Ultrasound (HIFU) has been verifiable by a change in average micelle diameter (from 26 nm to 90 nm). In addition, the authors confirmed that cleavage was superior to the triazole ring, at the central ester relation [132]. Pluronic F127 was recently used to create a new theranostic nanobubble (NB), which combines ultrasound and fluorescent pictorial tracking with Photodynamic therapy (PDT) [133].

Radiation responsive micelles

PDT uses chemical photosensitizer (PS), which, when irradiated with a particular wavelength, generates singlet oxygen and other ROS in tumor cells. During molecular interaction of active PS with intracellular oxygen, these radicals are formed. Tumor and healthy cells are classified by their metabolism. Increased metabolic and mitochondrial dysfunction allows cancer cells to exercise more oxidant stress through the oncogenic transition. Further anti-cancer effects of PDT occur by vascular shut-down, cell membrane de-stabilization, local immune system activation, and the injection of tumor antigen into infiltrating immune cells. Burst release and fast clearance of tumor area micelle can be overcome through the transformation of sole-to-gel [134-137].

Pluronic-based mixed polymeric micelles as drug solubilizing/ stabilizing carrier

The polymeric micelles allow the integration and protect against the inactivation of poorly water-soluble drugs into the biological media and strive to provide effective medication and targeting capabilities [135-138]. Several experiments have shown that the cytotoxicity of anti-cancer bioactive agents with MDR in tumor tissues can be improved by mixtures of pluronic and other polymers [116, 139, 140]. The self-assembly property of PF127 in the form of micelles was also assessed for the targeted supply of drugs. Nano-size pluronic micellar structures which be used to encapsulate hydrophobic agents on the surface of a nanoparticle inside the micelle's broad center or conjugate the hydrophilic moieties. It may also be used to grow hydrogels forming in situ, owing to thermo reacting features of the pluronic [141-145]. Hydrogels are ideal in situ for local as well as the systemic distribution of drugs. The pluronic agents are easily mixed, combined, or adsorbed by other common polymers such as chitosan, Polylactic Acid (PLA), PLGA, and so on in the field of drug delivery. Polymeric micelles are formed with lower levels and demonstrate greater thermodynamic and kinetic resilience relative to the micelles of standard surfactants to tolerate thermodynamic therapeutic dilution, as well as improved drug solubilization and stabilizing capacity [138, 146, 147]. Moreover, micelles in which hydrophilic blocks consist of PEO are sterically stabilized and macrophages are less feasible to consume [148]. Drug trapping in the micellar structure limits bond access to an external medium and thus, the drug-copolymer bond hydrolyze rate is much less than for the typical drug-polymer conjugates [149]. Only released drug molecules are anticipated to be pharmaceutical in a mechanically trapped environment, even though the micelle impacts cellular and body delivery [150].

The thermal transformation of Paclitaxel (PTX) to Pluronic-based NPs, with a core/shell configuration, was conducted in a mixture from the Pluronic F-68 to the liquid polyethylene glycol (PEG); molecular weight: 400). As PTX solubilizers, Liquid PEG and Pluronic F-68 are used for nanoencapsulation PEG containing PTX. In addition, emulsions made from PEG containing PTX and liquidized Pluronic F-68 were produced at nanometer level by extracting the melted mixture at a transition temperature (120 °C) as defined in fig. 5 [151]. The PEG containing PTX pluronic F-68 nanoencapsulation was finished with the liquid mixture being cooled to 0 °C. The formation of the PTX equipped Pluronic NPs with core/shell configuration has been clearly revealed by FE (field emission)-Scanning Electron Microscopy (SEM), cryo-transmitting electron microscopy (TEM), and by size distribution analysis. The sustained circulation in the bloodstream of the PTX-charged Pluronic NPs, resulting in improved tumor tissue targeting ability, was predicted to increase over that of the surfactant-based PTX, due to PEO blocks in pluronic F-68.

Fig. 5: The formation of Pluronic-based NPs with a core/shell structure. The core is composed of a mixture of PEG (molecular weight: 400) and PTX (or DTX) and the shell is composed of Pluronic F-68 [151]

Core/shell np composed of a pluronic composite

A temperature-induced phase change in the mixture between PLGA and Pluronic F-127 core/shell NPs with PLGA core and pluronic shells is prepared for a PTX carrier [152]. The liquidized mixtures of PLGA, PTX, and Pluronic F-127 were prepared at 600c on a stepped basis in response to temperature changes, with the result that the temperature was decreasing to 250C. Based on the actions of the PLGA and Pluronic F-127. SEM has been used to monitor the phase-separated state and check the identity of the PLGA center. When this mixture was spread into water, aqueous media is suppressed of the Pluronic-coated PLGA NPs (core/shell NPs with PLGA core). Fig. 6 explains the development of PLGA NPs with pluronic coating. On the surface of the PLGA heart, the pluronic shell tracked PTX from the core/shell NPs [153]. PLGA NPs were developed without the use of a poisonous organic solvent with the liquidized pluronic F-127 as a solvent. As the pluronic F-12, pluronic-coated PLGA NPs with a core/shell structure were not evaporated during preparation. The PTX and Docetaxel (DTX) models for small molecules were chosen and the vascular endothelial growth factor (VEGF) and HGH models were picked as protein-based medicines. They included both PTX and DTX. A pattern of continuous release of both model drugs has been found that the presence of a Pluronic coating on the liposome core surface mediated the release of the model drug.

Fig. 6: Temperature-induced phase transition in the melt mixture of PLGA and Pluronics [153]

Fig. 7: (a) Stabilization of Pluronic NPs by vesicle fusion (forming vesicular NPs) and (b) layer by layer approach stabilization of vesicle NPs [154]

Pluronic based NPs with a Core/Shell structure for cancer-targeting therapy

In the preparation of the NPs of Pluronic-based, temperature-induced phase transformation was very helpful. However, during the development of DTX charged Pluronic NPs during the previously mentioned temperature-induced transformation, the oxidation of DTX was observed [151]. The temperature-induced phase shift was rendered in a milder environment to protect DTX from oxidation (90 °C for 10 min). Though DTX oxidation was low, instability of DTX charged Pluronic NPs was observed with DTX precipitation within 10 min from the NPs in the aqueous medium.

The Pluronic NPs demonstrated a fast release pattern of precipitation of DTX in the release medium before being integrated into the vesicle [154]. In vesicle NPs, the release rate drop was controlled because the DTX released was penetrated through the lipid bilayer by the Pluronic NPs (fig. 7).

The multilayer NPs revealed a more deleted release rate for DTX as the external Pluronic layer was a further obstacle against DTX release. A calculation of anti-tumor effectiveness by tumor-bearing muzzles has observed the therapeutic functionality of the multilayer NPs. The DTX filled multi-layer NPs are more successful than those injecting free DTX (Commercial DTX (Taxotere®)), Empty NPs (Multilayer No-DTX), or Saline (200 L) [155, 156]. Due to their tumoral objective potential based on the EPR effect, the multilayer NP has demonstrated improved anti-tumor effectiveness [157]. This research prepared and injected multilayer NPs with iron oxide NPs in the tail veins of tumor-bearing mice to validate the ability of the multilayer NPs to target the tumor. There was a large improvement in MR strength at the tumor site relative to Resovist in the multi-layer iron oxide NPs. This shows that multilayer NPs can be used as nanocarriers with tumor-targeting capabilities for molecular imaging [158].

Building blocks for targeted chemotherapy using pluronics

The issues with conventional chemotherapy, such as a lack of precise targeting of the tumor and drug resistance, have been resolved with NP, whereby traditional chemotherapy has been improved [159-161]. Of these pluronic NPs, promising carriers have been identified in targeted cancer therapy. Due to Pluronics micellization in aquatic solution, polymeric micelles have been developed and their cores have been used as depots for different treatment agents and methods of diagnosis [162]. A Temperature Induced Phase Transition (TIPT), as defined in fig. 8, was prepared for pluronic NPs loaded by Paclitaxel (PTX). Images from Cryo TEM revealed Pluronic NPs' core/shell structure (fig. 8b).

Because the pluronic F-68 was mostly protected by the surface of NPs and PEOs, it was predicted that pluronic NPs would be retained in systemic circulation for a prolonged permeation and retention effect (EPR). This is the prerequisite. The high targeting efficiency near the tumor can be explained by the in vivo biodistribution in terms of the EPR effect. In Phase II clinical trials, esophageal adenocarcinoma was tested for the antitumor potency of the DOX formulation, composed from pluronic (L61 and F127) and DOX [163-165]. This technique was designed to test the multifunctional properties of the DOX-controlled pluronic/heparin-np-system. MB or DEVD-S-DOX was then achieved in the tumor-tissue mixture after photo-irradiation by the release of MB or DEVD-S-DOX from the NP (fig. 9). Table 4 illustrates several clinical trials involving Pluronic® polymer-containing formulations. Pluronic and heparin NPs were predicted to accumulate in tumor tissues caused by the EPR effect [166, 167]. Some medicines solubilized in poloxamer micelles used for cancer chemotherapy are presented in table 5 [71].

Fig. 8: (A) Formation of Pluronic nanoparticles by the TIPT method, (B) cryo-TEM image of paclitaxel-loaded Pluronic NPs [162]

Table 4: Some examples of pluronic®-containing formulations in clinical trials

Industry/sponsor Pluronic® containing formulation Use Stage Reference
Supratek Pharma Inc. SP1049C: Doxorubicin+Pluronics® L61 and F127 Advanced esophageal adenocarcinoma Phase III 168
Mast Therapeutics, Inc. Purified Poloxamer 188 (based on Pluronic® F68) Vaso-occlusive crisis Phase III
British Columbia Cancer Agency Topical amitriptyline 2%, ketamine1%, and lidocaine 5% in Pluronic® lecithin organogel Neuropathic pain secondary to radiation therapy

Phase III

Completed

CoDa Therapeutics, Inc. Nexagon®: Pluronic® gel Persistent corneal epithelial defects Phase II
Sancilio and Company, Inc. SC401B (Pluronic® F87 as a surfactant) Severe hypertriglyceridemia Phase III
Valentis VLTS-934 (Pluronic®Poloxamer 188) Peripheral vascular disease

Phase II

Completed

Pluronics as a therapeutic polymer

The biggest challenge for the success of chemical agents is tolerance. Resistance comes primarily from efflux pumps minimizing chemical therapeutic intracellular levels [169]. The therapeutic resistance induced by Efflux can be resolved by pluronic systems. Several studies record Pluronic alterations that induce decreased efflux activity in the lipid microenvironment of P-gp. The pluronic polymers suppress the membrane microviscosity that permeates the cancer cells to chemotherapeutic agents. Also, the mitochondrial membrane is destabilized by pluronic and induces significant ATP depletion in cancer cells. The Kabanov research group notes that pluronic can block the mitochondrial electron transfer chain (Complexes I and IV), which could increase the ROS level within the target cancer cells [170]. High ROS disturbs the mitochondrial membrane's normal configuration and initiates the release of apoptotic mediators Intrinsic [163, 171]. Pluronic cytochrome-mediated release c, Apoptosis-Inducing Factor (AIF), and Endonuclease G can cause other apoptotic mediators to be swiftly triggered within the programmed cell death protocol. Metastasis suppression is another essential function of the pluronic as a medicinal polymer. In metastatic cancers, the efficacy of normal medicinal substances is marginal. Pluronic polymers with medium hydrophilic-lipophilic equilibrium (HLB) have been documented to block the migration and invasion of cancer cells. In the 4T1 tumor-carrying mice model, a major inhibition of the lung metastasis by a pluronic effect was reported [172]. Oddly, pluronic Antimetastatic characteristics are linked to a decrease in matrix metaloproteases (MMP) such as MMP-9 control. Moreover, the above-mentioned study has documented substantial synergistic anti-metastatic activity for the combination of pluronic-doxorubicin. In another related analysis, the P85 pluronic copolymer micelles loaded with a 5-Fluorouracil (5FU) were produced for colon cancer inhibition [173]. There has recently been a new pluronic F68 conjugated delivery system [174]. The potency and toxicity of these micelles concerning naked drugs were appropriate. Gemcitabine is a 70-gent anti-cancer drug used in multiple chemo regimes. Dinarv and the study community reveal a novel chitosan-pluronic oral delivery system. Pluronic nanoparticles hydrophilic shells can be used to conjugate target moieties such as folates, aptamers, and monoclonal antibodies (fig. 10). There is numerous folate-conjugated pluronic framework for the transmission of paclitaxel and doxorubicin, which has shown substantial tumor aggregation folate cross-connected pluronic micelles [175, 176]. Rabial Glycoprotein Virus (RVG) conjugates pluronic nanoparticles is another groundbreaking nanocarrier. RVG has effectively conjugated particles aim at the blood-brain barrier and has thus increased the biological bioavailability of the brain. The treatment of different brain tumors or Central Nervous System (CNS) malignancies may be achieved with such a method. Pluronic supply systems could be used as a vehicle for the supply of combinatorial regimens because of versatile and tunable physicochemical properties [177]. Paclitaxel-loaded pluronic Arginyl Glycylaspartic Acid (RGD)-plated nanoparticles for the treatment of gliomas have been analyzed in another report [178]. RGD peptide has been used to cross the blood-brain barrier as a penetration enhancer.

Fig. 9: An illustrative description of chemo-photodynamic combination therapy [76, 119]

Table 5: Anticancer agents formulated in poloxamer-based polymeric micelles

Drug Copolymer Performance Reference
Epirubicin L61, P85, F108 Lifespan of animals and inhibition of tumor growth considerably increased with drug/copolymer compositions. 71
Doxorubicin P105 Lower in vitro proliferation of MatLu rat prostate carcinoma cells with micellar system.
P85 A formulation containing Pluronic® P85 and DOX prevents the development of MDR in the MCF7 human breast carcinoma cell line.
Camptothecin F127, L92 versus materials modified with Polyacrylic acid (PAA) blocks 3-to 4-fold higher solubility. Amount of camptothecin solubilized per PPO greater in the Pluronic-PAA than the parent, suggesting solubilization by the hydrophobic cores and hydrophilic shells. Enhanced stability.
F127, F68, P85 Improved oral absorption estimated in vitro.
Megestrol F127/l61 mixed micelles versus materials modified with PAA blocks Bioadhesive tablets for gastrointestinal retention and enhanced bioavailability.
Paclitaxel P123 Enhanced solubility, prolonged blood circulation and modified biodistribution. Plasma half-life was 2.3-fold higher. Increased accumulation of PTX in ovary, uterus, lung, and kidney; but decreased accumulation in liver and brain.
Octaethylporphine F127, F68, P85 Improved oral absorption estimated in vitro.
Epidermal growth factor F127 Gel was evaluated as a potential topical vehicle.
Tyrphostin 47 F127 Sustained local delivery does not result in a reduction of neointimal proliferation in the rat carotid injury model.
Rapamycin F127 Treatment of experimental vein grafts is associated to increased apoptosis in the vascular wall and reduction of neointimal hyperplasia.
Methotrexate F127 Potential direct administration into solid tumors

Fig. 10: Pluronic based delivery system; a novel platform for formulation of hydrophobic molecules and targeted delivery [174]

Pluronic nanocarrier: a theranostic approach

Nanoscale theranostic such as polymeric micelles are commonly explored as promising gold standards in a personal medicine context for the purpose of diagnosis, treat and track the growth of tumors simultaneously [179]. Theranostic functional pluronic polymer micelles have shown tremendous promise in contrast to traditional therapies to enhance and track medication delivery after administration, which can improve drug effectiveness and mitigate off-target toxicity. Latest studies have indicated that the tumor microenvironment (TME), including malignancy, invasion, and metastasis, is a central orchestrator of cancer progression [180, 181]. A significant biological factor in which cohesive and polarized epithelial cells turn over nonpolarized and highly mobile, mesenchymal-like cells remains an important element of epithelial-mesenchymal transfer [182]. The innovative design of nanomedicine continues to be relevant as the tumor barriers to drug accumulation (altered flux, thick matrix, efflux pumps.) are multiple and highly effective. In particular, the use of nano-caregiators, adorned with the most adequate targeting vector and with the most suitable therapeutic agent, all selected in the light of genetic knowledge for the recipient, is a means of successful transmission to the cells involved, while the protection and efficacy of the therapy are far-reaching [183, 184]. The hydrated pluronic PEO shell induces sterical repulsion and leads to a strongly activated protein adsorption strength barrier [185, 186]. Some examples of pluronic micelles for cancer diagnosis are been listed in table 6. It thus minimizes the formation of protein corona that contributes to the prolonged blood circulation and bio-imaging performance of the nanocarrier for theranostic (fig. 11) [187].

The Pluronic P94 was studied to direct the intravenous and intratumoral injections of radionuclides [188]. In for the hybrid Single-Photon Emission Computerized Tomography (SPECT)/ Computed Tomography (CT) imaging pluronic F68 micelles incorporating Near-Infrared (NIR) Cy 5.5 and DOX is found useful in the diagnosis and treatment of targets. Pluronic F127 is safe against protein adsorption. Table No.6 outlines recent techniques for multi-factor therapies, medications, and/or theranostic of cancer, including Pluronic structures alone and as a mixed method. In combination with biomethodologies, pluronic intelligent nano micelles will open the door for understanding the pathways of cancer that are at the core of the diseases, including the Epithelial-Mesenchymal Transition (EMT)-related methods.

Table 6: Overview of some pluronic micelles for cancer diagnosis; theranostic approach

Pluronic Imaging
Agent

Modality

Drug Theranostic model Result Purpose Reference
Pluronic F68 Cyanine5.5 (Cy5.5) dye NIR DTX HIFU

In vitro SCC-7 cells, murine

in vivo male C3H/HeN mice

Targetable/ Triggering nanosystems for the solid tumors.

PMs triggering release into tumor cells occurs under HIFU exposure through nonthermal mechanisms, which increase the therapeutic effect.

189
F127 β-thiophene-fused-BF2 azadi-pyrromethene (aza BDTP) NIR PTX Photoacoustic imaging and photothermal

In vitro 4T1 cells, mouse

breast cancer cell line

Co-loading of aza-BDTP and PTX (BDTP/PTX micelles show promising in vitro/in vivo results as nanotheranostic vehicles.) 190
F127 AuNPs NIR PTX

chemo-photothermal

therapy

in vitro MDA-MB-231 cells

in vivo female Balb/c nude

In vitro and in vivo studies using the combined strategy. (The pluronic-PLL-Au micellar carrier can cause a synergistic effect that is promising for chemo-photothermal therapy.) 191
F127 AuNPs IR780 iodide NIR105 IR780 photodynamic and photo-thermal therapies and surface-enhanced reso-nance Raman scattering in vitro C26 cells, murine colon carcinoma cell line The use of the AuNPs-F127-IR780 micellar system indicates synergistic effects by simultaneous photodynamic and photothermal activity. 191
P123 F127 rhodamine-B dye NIR Verte-porfin Photodynamic therapy in vitro MCF-7 cells, human breast cancer cell line PC3 cells, human prostate cancer cell line Multifunctional pluronic P123/F127 mixed micelles show promising results for the encapsulation and delivery of the photodynamic therapy. 192

Fig. 11: Multifunctional theranostic and bio imaging nanosystems. PET, positron-electron transmission; SPECT, single-photon emission computed tomography; CT, computed tomography; MRI, magnetic resonance imaging; NPs, nanoparticles; US, ultrasounds; OI, optical imaging [187]

Future prospective

In clinical trials as vehicles of cancer therapies, there are some promising practical pluronic micelle formulations. The clinical translation of the nano-medicines is still a problem. However, their use in theranostic has not yet been translated into clinical trials [193]. The copolymer concentration can decay below the CMC, for example, after intravenous administration and subsequent formulation dilution, resulting in micellar dissociations and premature release of medicines. Polymer micelles must also be programmed for extended blood circulation to ensure a proper concentration of the loaded therapeutic product and the photographic material collecting in the target area. While it is possible to use multi-target nano delivery schemes, the still enormous distance between preclinical and clinical outcomes requires the introduction of general and logical translation protocols [194-197]. The main concern of future research can be done in the preparation of nanoparticles that can further withstand the biological diversities and thus further improve drug stability in the biological environment and hence its bioavailability [198].

CONCLUSION

Conventional cytotoxic medications induce some side effects partly as a result of existing treatment protocols dependent on several administrative periods. The effectiveness of the strategy is dependent on the combination of recent trends in nanomedicine care with targeted cytotoxic drug nanocarrier allowing for sustained release, site-specific delivery, and dissemination, reduction, or even elimination. One of the most brilliant cancer therapy techniques has been the advancement of nanoparticles-based treatment strategies as a drug delivery method. The conjugation of ligands on the surface of nanoparticles for cell recognition has resulted in the development of a new generation of nanoparticles (targeted nanoparticles). The use of differentially expressing molecules such as polymeric nanoparticles on the surrounding tumors could be achieved in the targeted nanoparticles offering sufficient cytotoxic release. The use of tailored and functionalized NPs promises to advance in emerging therapy groups. A multifunctional theranostic approach in the drug delivery system can be successful against cancer with different therapeutic cargo. These forms of formulations can be investigated in clinical trials.

FUNDING

Nil

AUTHORS CONTRIBUTIONS

All the authors have contributed equally.

CONFLICTS OF INTERESTS

Declared none

REFERENCES

  1. Pucci C, Martinelli C, Ciofani G. Innovative approaches for cancer treatment: current perspectives and new challenges. Ecancermedicalscience. 2019;13:961. doi: 10.3332/ecancer.2019.961, PMID 31537986.

  2. Wadhwa A, Mathura V, Lewis SA. Emerging novel Nano pharmaceuticals for drug delivery. Asian J Pharm Clin Res. 2018;11(7):35-42. doi: 10.22159/ajpcr.2018.v11i7.25149.

  3. Jahan ST, Sadat SMA, Walliser M, Haddadi A. Targeted therapeutic nanoparticles: an immense promise to fight against cancer. J Drug Delivery. 2017;2017:9090325. doi: 10.1155/2017/9090325, PMID 29464123.

  4. Urruticoechea A, Alemany R, Balart J, Villanueva A, Vinals F, Capella G. Recent advances in cancer therapy: an overview. Curr Pharm Des. 2010;16(1):3-10. doi: 10.2174/138161210789941847, PMID 20214614.

  5. Williams GH, Stoeber K. The cell cycle and cancer. J Pathol. 2012;226(2):352-64. doi: 10.1002/path.3022, PMID 21990031.

  6. Meyers CA. How chemotherapy damages the central nervous system. J Biol. 2008;7(4):11. doi: 10.1186/jbiol73, PMID 18439322.

  7. Ismaili N, Amzerin M, Flechon A. Chemotherapy in advanced bladder cancer: current status and future. J Hematol Oncol. 2011;4(1):35. doi: 10.1186/1756-8722-4-35.

  8. DeVita VT, Chu E. A history of cancer chemotherapy. Cancer Res. 2008;68(21):8643-53. doi: 10.1158/0008-5472.CAN-07-6611, PMID 18974103.

  9. Washington CM, Leaver DT. Principles and practice of radiation therapy-e-book. Elsevier Health Sciences; 2015.

  10. Baskar R, Lee KA, Yeo R, Yeoh KW. Cancer and radiation therapy: current advances and future directions. Int J Med Sci. 2012;9(3):193-9. doi: 10.7150/ijms.3635, PMID 22408567.

  11. Naef AP. Hugh morriston davies: first dissection lobectomy in 1912. Ann Thorac Surg. 1993;56(4):988-9. doi: 10.1016/0003-4975(93)90377-t, PMID 8215687.

  12. Phillips EH, Franklin M, Carroll BJ, Fallas MJ, Ramos R, Rosenthal D. Laparoscopic colectomy. Ann Surg. 1992;216(6):703-7. doi: 10.1097/00000658-199212000-00015, PMID 1466626.

  13. Singletary SE. Minimally invasive techniques in breast cancer treatment. Semin Surg Oncol. 2001;20(3):246-50. doi: 10.1002/ssu.1040, PMID 11523110.

  14. Sherwood JT, Brock MV. Lung cancer: new surgical approaches. Respirology. 2007;12(3):326-32. doi: 10.1111/j.1440-1843.2007.01083.x, PMID 17539834.

  15. Hashizume M. MRI-guided laparoscopic and robotic surgery for malignancies. Int J Clin Oncol. 2007;12(2):94-8. doi: 10.1007/s10147-007-0664-z, PMID 17443276.

  16. Sun C, Fang C, Stephen Z, Veiseh O, Hansen S, Lee D, Ellenbogen RG, Olson J, Zhang M. Tumor-targeted drug delivery and MRI contrast enhancement by chlorotoxin-conjugated iron oxide nanoparticles. Nanomedicine (Lond). 2008;3(4):495-505. doi: 10.2217/17435889.3.4.495, PMID 18694312.

  17. Chertok B, Moffat BA, David AE, Yu F, Bergemann C, Ross BD, Yang VC. Iron oxide nanoparticles as a drug delivery vehicle for MRI monitored magnetic targeting of brain tumors. Biomaterials. 2008;29(4):487-96. doi: 10.1016/j.biomaterials. 2007.08.050, PMID 17964647.

  18. Dhar S, Gu FX, Langer R, Farokhzad OC, Lippard SJ. Targeted delivery of cisplatin to prostate cancer cells by aptamer functionalized Pt(IV) prodrug-PLGA-PEG nanoparticles. Proc Natl Acad Sci USA. 2008;105(45):17356-61. doi: 10.1073/pnas.0809154105, PMID 18978032.

  19. Fayter D, Corbett M, Heirs M, Fox D, Eastwood A. A systematic review of photodynamic therapy in the treatment of pre-cancerous skin conditions, Barrett’s oesophagus and cancers of the biliary tract, brain, head and neck, lung, oesophagus and skin. In: NIHR Health Technology Assessment Programme: executive summaries. NIHR J Library; 2010.

  20. Huang X, Jain PK, El-Sayed IH, El-Sayed MA. Plasmonic photothermal therapy (PPTT) using gold nanoparticles. Lasers Med Sci. 2008;23(3):217-28. doi: 10.1007/s10103-007-0470-x, PMID 17674122.

  21. Griffin RJ, Dings RP, Jamshidi-Parsian A, Song CW. Mild temperature hyperthermia and radiation therapy: role of tumour vascular thermotolerance and relevant physiological factors. Int J Hyperthermia. 2010;26(3):256-63. doi: 10.3109/02656730903453546, PMID 20210610.

  22. Hurwitz MD. Today’s thermal therapy: not your father’s hyperthermia: challenges and opportunities in the application of hyperthermia for the 21st-century cancer patient. Am J Clin Oncol. 2010;33(1):96-100. doi: 10.1097/COC.0b013e3181817a75, PMID 19636240.

  23. Jordan A, Scholz R, Maier-Hauff K, van Landeghem FK, Waldoefner N, Teichgraeber U, Pinkernelle J, Bruhn H, Neumann F, Thiesen B, von Deimling A, Felix R. The effect of thermotherapy using magnetic nanoparticles on rat malignant glioma. J Neurooncol. 2006;78(1):7-14. doi: 10.1007/s11060-005-9059-z, PMID 16314937.

  24. Kawai N, Ito A, Nakahara Y, Futakuchi M, Shirai T, Honda H, Kobayashi T, Kohri K. Anticancer effect of hyperthermia on prostate cancer mediated by magnetite cationic liposomes and immune‐response induction in transplanted syngeneic rats. Prostate. 2005;64(4):373-81. doi: 10.1002/pros.20253, PMID 15754344.

  25. Maier Hauff K, Rothe R, Scholz R, Gneveckow U, Wust P, Thiesen B, Feussner A, von Deimling A, Waldoefner N, Felix R, Jordan A. Intracranial thermotherapy using magnetic nanoparticles combined with external beam radiotherapy: results of a feasibility study on patients with glioblastoma multiforme. J Neurooncol. 2007;81(1):53-60. doi: 10.1007/ s11060-006-9195-0, PMID 16773216.

  26. Lehmann J, Natarajan A, DeNardo GL, Ivkov R, Foreman AR, Catapano C, Mirick G, Quang T, Gruettner C, Denardo SJ. Short communication: nanoparticle thermotherapy and external beam radiation therapy for human prostate cancer cells. Cancer Biother Radiopharm. 2008;23(2):265-71. doi: 10.1089/ cbr.2007.0411, PMID 18454696.

  27. Dudley ME, Yang JC, Sherry R, Hughes MS, Royal R, Kammula U, Robbins PF, Huang J, Citrin DE, Leitman SF, Wunderlich J, Restifo NP, Thomasian A, Downey SG, Smith FO, Klapper J, Morton K, Laurencot C, White DE, Rosenberg SA. Adoptive cell therapy for patients with metastatic melanoma: evaluation of intensive myeloablative chemoradiation preparative regimens. J Clin Oncol. 2008;26(32):5233-9. doi: 10.1200/ JCO.2008.16.5449, PMID 18809613.

  28. Rosenberg SA, Dudley ME. Adoptive cell therapy for the treatment of patients with metastatic melanoma. Curr Opin Immunol. 2009;21(2):233-40. doi: 10.1016/j.coi.2009.03.002, PMID 19304471.

  29. Ren J, Di L, Song G, Yu J, Jia J, Zhu Y, Yan Y, Jiang H, Liang X, Che L, Zhang J, Wan F, Wang X, Zhou X, Lyerly HK. Selections of an appropriate regimen of high-dose chemotherapy combined with adoptive cellular therapy with dendritic and cytokine-induced killer cells improved progression-free and overall survival in patients with metastatic breast cancer: reargument of such contentious therapeutic preferences. Clin Transl Oncol. 2013;15(10):780-8. doi: 10.1007/s12094-013-1001-9, PMID 23359185.

  30. Arruebo M, Vilaboa N, Saez-Gutierrez B, Lambea J, Tres A, Valladares M, González-Fernández A. Assessment of the evolution of cancer treatment therapies. Cancers. 2011;3(3):3279-330. doi: 10.3390/cancers3033279, PMID 24212956.

  31. Patil PM, Chaudhari PD, Sahu M, Duragkar NJ. Review article on gene therapy. Res J Pharmacol Pharmacodyn. 2012;4:77-83.

  32. Sanna V, Pala N, Sechi M. Targeted therapy using nanotechnology: focus on cancer. Int J Nanomedicine. 2014;9:467-83. doi: 10.2147/IJN.S36654, PMID 24531078.

  33. Cisterna BA, Kamaly N, Choi WI, Tavakkoli A, Farokhzad OC, Vilos C. Targeted nanoparticles for colorectal cancer. Nanomedicine (Lond). 2016;11(18):2443-56. doi: 10.2217/nnm-2016-0194, PMID 27529192.

  34. Yuan X, Kang C, Zhao Y, Gu M, Pu P, Tian N, Sheng J. Surface multi-functionalization of poly (lactic acid) nanoparticles and c6 glioma cell targeting in vivo. Chinese J Polym Sci. 2009;27(2):231-9. doi: 10.1142/S0256767909003868.

  35. Zhou J, Romero G, Rojas E, Ma L, Moya S, Gao C. Layer by layer chitosan/alginate coatings on poly (lactide-co-glycolide) nanoparticles for antifouling protection and folic acid-binding to achieve selective cell targeting. J Colloid Interface Sci. 2010;345(2):241-7. doi: 10.1016/j.jcis.2010.02.004, PMID 20227712.

  36. Li Z, Tan S, Li S, Shen Q, Wang K. Cancer drug delivery in the Nano era: an overview and perspectives. Oncol Rep. 2017;38(2):611-24. doi: 10.3892/or.2017.5718, PMID 28627697.

  37. Hemant K, Raizaday A, Sivadasu P, Uniyal S, Kumar SH. Cancer nanotechnology: nanoparticulate drug delivery for the treatment of cancer. Int J Pharm Pharm Sci. 2015;7:40-6.

  38. Autio KA, Garcia JA, Alva AS, Hart LL, Milowsky MI, Posadas EM. A phase 2 study of BIND-014 (PSMA-targeted docetaxel nanoparticle) was administered to patients with chemotherapy-naïve metastatic castration-resistant prostate cancer (mCRPC). American Society of Clinical Oncology; 2016.

  39. Elsabahy M, Wooley KL. Design of polymeric nanoparticles for biomedical delivery applications. Chem Soc Rev. 2012;41(7):2545-61. doi: 10.1039/c2cs15327k, PMID 22334259.

  40. Mohamed F, van der Walle CF. Engineering biodegradable polyester particles with specific drug targeting and drug release properties. J Pharm Sci. 2008;97(1):71-87. doi: 10.1002/jps.21082, PMID 17722085.

  41. Danhier F, Feron O, Préat V. To exploit the tumor microenvironment: passive and active tumor targeting of nanocarriers for anti-cancer drug delivery. J Control Release. 2010;148(2):135-46. doi: 10.1016/j.jconrel.2010.08.027, PMID 20797419.

  42. Pelicano H, Martin DS, Xu RH, Huang P. Glycolysis inhibition for anticancer treatment. Oncogene. 2006;25(34):4633-46. doi: 10.1038/sj.onc.1209597, PMID 16892078.

  43. Bazak R, Houri M, El Achy SE, Hussein W, Refaat T. Passive targeting of nanoparticles to cancer: a comprehensive review of the literature. Mol Clin Oncol. 2014;2(6):904-8. doi: 10.3892/mco.2014.356, PMID 25279172.

  44. Cengelli F, Maysinger D, Tschudi Monnet F, Montet X, Corot C, Petri Fink A, Hofmann H, Juillerat Jeanneret L. Interaction of functionalized superparamagnetic iron oxide nanoparticles with brain structures. J Pharmacol Exp Ther 2006;318(1):108-16. doi: 10.1124/jpet.106.101915, PMID 16608917.

  45. Jokerst JV, Lobovkina T, Zare RN, Gambhir SS. Nanoparticle PEGpegylation for imaging and therapy. Nanomedicine (Lond). 2011;6(4):715-28. doi: 10.2217/nnm.11.19, PMID 21718180.

  46. Moghimi SM, Hunter AC. Poloxamers and poloxamines in nanoparticle engineering and experimental medicine. Trends Biotechnol. 2000;18(10):412-20. doi: 10.1016/s0167-7799(00)01485-2, PMID 10998507.

  47. Shirshahi V, Shamsipour F, Zarnani AH, Verdi J, Saber R. Active targeting of HER2-positive breast cancer cells by Herceptin-functionalized organically modified silica nanoparticles. Cancer Nanotechnol. 2013;4(1-3):27-37. doi: 10.1007/s12645-013-0035-6, PMID 26069499.

  48. Yang Z, Kang SG, Zhou R. Nanomedicine: de novo design of nanodrugs. Nanoscale. 2014;6(2):663-77. doi: 10.1039/c3nr04535h, PMID 24305636.

  49. Muro S. Challenges in design and characterization of ligand-targeted drug delivery systems. J Controlled Release. 2012;164(2):125-37. doi: 10.1016/j.jconrel.2012.05.052, PMID 22709588.

  50. Wang J, Tian S, Petros RA, Napier ME, DeSimone JM. The complex role of multivalency in nanoparticles targeting the transferrin receptor for cancer therapies. J Am Chem Soc. 2010;132(32):11306-13. doi: 10.1021/ja1043177, PMID 20698697.

  51. Lee H, Fonge H, Hoang B, Reilly RM, Allen C. The effects of particle size and molecular targeting on the intratumoral and subcellular distribution of polymeric nanoparticles. Mol Pharm. 2010;7(4):1195-208. doi: 10.1021/mp100038h, PMID 20476759.

  52. Xin H, Jiang X, Gu J, Sha X, Chen L, Law K, Chen Y, Wang X, Jiang Y, Fang X. Angiopep-conjugated poly (ethylene glycol)-co-poly (ε-caprolactone) nanoparticles as dual-targeting drug delivery system for brain glioma. Biomaterials. 2011;32(18):4293-305. doi: 10.1016/j.biomaterials.2011.02.044, PMID 21427009.

  53. Kim BJ, Im SS, Oh SG. Investigation on the solubilization locus of aniline-HCl salt in SDS micelles with 1H NMR spectroscopy. Langmuir. 2001;17(2):565-6. doi: 10.1021/la0012889.

  54. Sun B, Ranganathan B, Feng SS. Multifunctional poly (D, L-lactide-co-glycolide)/montmorillonite (PLGA/MMT) nanoparticles decorated by Ttrastuzumab for targeted chemotherapy of breast cancer. Biomaterials. 2008; 29(4):475-86. doi: 10.1016/j.biomaterials.2007.09.038, PMID 17953985.

  55. Cruz LJ, Tacken PJ, Fokkink R, Joosten B, Stuart MC, Albericio F, et al, Torensma R, Figdor CG. Targeted PLGA nNano-but not microparticles specifically deliver antigen to human dendritic cells via DC-SIGN in vitro. J Controlled Release. 2010;144(2):118-26. doi: 10.1016/j.jconrel.2010.02.013.

  56. Dhar S, Gu FX, Langer R, Farokhzad OC, Lippard SJ. Targeted delivery of cisplatin to prostate cancer cells by aptamer functionalized Pt(IV) prodrug-PLGA–PEG nanoparticles. Proc Natl Acad Sci USA. 2008;105(45):17356-61. doi: 10.1073/pnas.0809154105, PMID 18978032.

  57. Torchilin V. Tumor delivery of macromolecular drugs based on the EPR effect. Adv Drug Delivery Rev. 2011;63(3):131-5. doi: 10.1016/j.addr.2010.03.011, PMID 20304019.

  58. Wang Z, Chui WK, Ho PC. Design of a multifunctional PLGA nanoparticulate drug delivery system: evaluation of its physicochemical properties and anticancer activity to malignant cancer cells. Pharm Res. 2009;26(5):1162-71. doi: 10.1007/s11095-009-9837-y, PMID 19191012.

  59. Farokhzad OC, Cheng J, Teply BA, Sherifi I, Jon S, Kantoff PW, Richie JP, Langer R. Targeted nanoparticle-aptamer bioconjugates for cancer chemotherapy in vivo. Proc Natl Acad Sci USA. 2006;103(16):6315-20. doi: 10.1073/ pnas.0601755103, PMID 16606824.

  60. Esmaeili F, Ghahremani MH, Ostad SN, Atyabi F, Seyedabadi M, Malekshahi MR, Amini M, Dinarvand R. Folate-receptor-targeted delivery of docetaxel nanoparticles prepared by PLGA–PEG–folate conjugate. J Drug Target. 2008;16(5):415-23. doi: 10.1080/10611860802088630, PMID 18569286.

  61. Tiwari S, Kansara V, Bahadur P. Targeting anticancer drugs with pluronic aggregates: recent updates. Int J Pharm. 2020;586:119544. doi: 10.1016/j.ijpharm.2020.119544.

  62. Akash MSH, Rehman K. Recent progress in biomedical applications of pluronic (PF127): pharmaceutical perspectives. J Controlled Release. 2015;209:120-38. doi: 10.1016/j.jconrel.2015.04.032, PMID 25921088.

  63. Bodratti AM, Sarkar B, Alexandridis P. Adsorption of poly (ethylene oxide)-containing amphiphilic polymers on solid–liquid interfaces: fundamentals and applications. Adv Colloid Interface Sci. 2017;244:132-63. doi: 10.1016/j.cis.2016.09.003, PMID 28069108.

  64. Lahiri B, Mukhopadhyay SD. Credibility of farm information disseminated through newspapers and radio programme: a case study. Indian Res J Ext Educ. 2016;13:1-8.

  65. Yang L, Alexandridis P. Physicochemical aspects of drug delivery and release from polymer-based colloids. Curr Opin Colloid Interface Sci. 2000;5(1-2):132-43. doi: 10.1016/S1359-0294(00)00046-7.

  66. Alexandridis P. Gold nanoparticle synthesis, morphology control, and stabilization facilitated by functional polymers. Chem Eng Technol. 2011;34(1):15-28. doi: 10.1002/ceat.201000335.

  67. Agnely F, Djedour A, Bochot A, Grossiord JL. Properties of various thermoassociating polymers: pharmaceutical and cosmetic applications. J Drug Delivery Sci Technol. 2006;16(1):3-10. doi: 10.1016/S1773-2247(06)50001-2.

  68. Tadros T. Viscoelastic properties of sterically stabilised emulsions and their stability. Adv Colloid Interface Sci. 2015;222:692-708. doi: 10.1016/j.cis.2015.03.001, PMID 25900262.

  69. Tadros TF. Interfacial phenomena and colloid stability: industrial applications. Walter de Gruyter GmbH & Co KG; 2015.

  70. Chang Y, Chu WL, Chen WY, Zheng J, Liu L, Ruaan RC. A systematic SPR study of human plasma protein adsorption behavior on the controlled surface packing of self‐assembled poly (ethylene oxide) triblock copolymer surfaces. J Biomed Mater Res Part. 2010;93:400-8.

  71. Alvarez Lorenzo C, Sosnik A, Concheiro A. PEO-PPO block copolymers for passive micellar targeting and overcoming multidrug resistance in cancer therapy. Curr Drug Targets. 2011;12(8):1112-30. doi: 10.2174/138945011795906615, PMID 21443477.

  72. Singh Joy SD, McLain VC. Safety assessment of poloxamers 101, 105, 108, 122, 123, 124, 181, 182, 183, 184, 185, 188, 212, 215, 217, 231, 234, 235, 237, 238, 282, 284, 288, 331, 333, 334, 335, 338, 401, 402, 403, and 407, poloxamer 105 benzoate, and poloxamer 182 dibenzoate as used in cosmetics. Int J Toxicol. 2008;27 Suppl 2:93-128. doi: 10.1080/10915810802244595, PMID 18830866.

  73. Grindel JM, Jaworski T, Piraner O, Emanuele RM, Balasubramanian M. Distribution, metabolism, and excretion of a novel surface-active agent, purified poloxamer 188, in rats, dogs, and humans. J Pharm Sci. 2002;91(9):1936-47. doi: 10.1002/jps.10190, PMID 12210041.

  74. Batrakova E, Lee S, Li S, Venne A, Alakhov V, Kabanov A. Fundamental relationships between the composition of pluronic block copolymers and their hypersensitization effect in MDR cancer cells. Pharm Res. 1999;16(9):1373-9. doi: 10.1023/a:1018942823676, PMID 10496652.

  75. Venne A, Li S, Mandeville R, Kabanov A, Alakhov V. Hypersensitizing effect of pluronic L61 on cytotoxic activity, transport, and subcellular distribution of doxorubicin in multiple drug-resistant cells. Cancer Res. 1996;56(16):3626-9. PMID 8705995.

  76. Kabanov AV, Batrakova EV, Alakhov VY. Pluronic block copolymers for overcoming drug resistance in cancer. Adv Drug Delivery Rev. 2002;54(5):759-79. doi: 10.1016/s0169-409x(02)00047-9, PMID 12204601.

  77. Altan N, Chen Y, Schindler M, Simon SM. Defective acidification in human breast tumor cells and implications for chemotherapy. J Exp Med. 1998;187(10):1583-98. doi: 10.1084/jem.187.10.1583, PMID 9584137.

  78. Breuninger LM, Paul S, Gaughan K, Miki T, Chan A, Aaronson SA, Kruh GD. Expression of the multidrug resistance-associated protein in NIH/3T3 cells confers multidrug resistance associated with increased drug efflux and altered intracellular drug distribution. Cancer Res. 1995;55(22):5342-7. PMID 7585598.

  79. Cleary I, Doherty G, Moran E, Clynes M. The multidrug-resistant human lung tumour cell line, DLKP-A10, expresses novel drug accumulation and sequestration systems. Biochem Pharmacol. 1997;53(10):1493-502. doi: 10.1016/s0006-2952(97)00003-8, PMID 9260877.

  80. Nooter K, Stoter G. Molecular mechanisms of multidrug resistance in cancer chemotherapy. Pathol Res Pract. 1996;192(7):768-80. doi: 10.1016/S0344-0338(96)80099-9, PMID 8880878.

  81. Shapiro AB, Fox K, Lee P, Yang YD, Ling V. Functional intracellular P‐glycoprotein. Int J Cancer. 1998;76(6):857-64. doi: 10.1002/(sici)1097-0215(19980610)76:6<857:aid-ijc15>3.0.co;2-#, PMID 9626353.

  82. Benderra Z, Morjani H, Trussardi A, Manfait M. Role of the vacuolar H+-ATPase in daunorubicin distribution in etoposide-resistant MCF7 cells overexpressing the multidrug-resistance associated protein. Int J Oncol. 1998;12(3):711-5. doi: 10.3892/ijo.12.3.711, PMID 9472114.

  83. Jung YW, Lee H, Kim JY, Koo EJ, Oh KS, Yuk SH. Pluronic-based core/shell nanoparticles for drug delivery and diagnosis. Curr Med Chem. 2013;20(28):3488-99. doi: 10.2174/09298673113209990036, PMID 23745558.

  84. Aydin F, Chu X, Uppaladadium G, Devore D, Goyal R, Murthy NS, Zhang Z, Kohn J, Dutt M. Self-assembly and critical aggregation concentration measurements of ABA triblock copolymers with varying B block types: model development, prediction, and validation. J Phys Chem B. 2016;120(15):3666-76. doi: 10.1021/acs.jpcb.5b12594, PMID 27031284.

  85. Srinivas G, Discher DE, Klein ML. Self-assembly and properties of diblock copolymers by coarse-grain molecular dynamics. Nat Mater. 2004;3(9):638-44. doi: 10.1038/nmat1185, PMID 15300242.

  86. Nawaz S, Carbone P. Coarse-graining poly (ethylene oxide)–poly (propylene oxide)–poly (ethylene oxide) (PEO–PPO–PEO) block copolymers using the MARTINI force field. J Phys Chem B. 2014;118(6):1648-59. doi: 10.1021/jp4092249, PMID 24446682.

  87. Bressel K, Gradzielski M. Enhancing the stability of spontaneously self-assembled vesicles - the effect of polymer architecture. Soft Matter. 2015;11(12):2445-53. doi: 10.1039/c4sm02746a, PMID 25668397.

  88. Bryskhe K, Jansson J, Topgaard D, Schillén K, Olsson U. Spontaneous vesicle formation in a block copolymer system. J Phys Chem B. 2004;108(28):9710-9. doi: 10.1021/jp031313u.

  89. Nagarajan R. ’Non-equilibrium’ block copolymer micelles with glassy cores: A predictive approach based on the theory of equilibrium micelles. J Colloid Interface Sci. 2015;449:416-27. doi: 10.1016/j.jcis.2014.12.077, PMID 25595626.

  90. Bouchemal K, Agnely F, Koffi A, Ponchel G. A concise analysis of the effect of temperature and propanediol-1, 2 on pluronic F127 micellization using isothermal titration microcalorimetry. J Colloid Interface Sci. 2009;338(1):169-76. doi: 10.1016/j.jcis.2009.05.075, PMID 19580975.

  91. Liang X, Guo C, Ma J, Wang J, Chen S, Liu H. Temperature-dependent aggregation and disaggregation of poly (ethylene oxide)-poly (propylene oxide)-poly (ethylene oxide) block copolymer in aqueous solution. J Phys Chem B. 2007;111(46):13217-20. doi: 10.1021/jp074990n, PMID 17973418.

  92. Ganguly R, Aswal VK, Hassan PA, Gopalakrishnan IK, Yakhmi JV. Sodium chloride and ethanol-induced sphere to rod transition of triblock copolymer micelles. J Phys Chem B. 2005;109(12):5653-8. doi: 10.1021/jp0468408, PMID 16851610.

  93. Narang P, Yadav N, Venkatesu P. Scrutinizing the effect of various nitrogen-containing additives on the micellization behavior of a triblock copolymer. J Colloid Interface Sci. 2019;553:655-65. doi: 10.1016/j.jcis.2019.06.074, PMID 31252181.

  94. Parekh P, Dey J, Kumar S, Nath S, Ganguly R, Aswal VK, Bahadur P. Butanol solubilization in aqueous F127 solution: investigating the enhanced micellar solvation and consequent improvement in gelation characteristics. Colloids Surf B Biointerfaces. 2014;114:386-91. doi: 10.1016/j.colsurfb.2013.10.030, PMID 24252230.

  95. Bharatiya B, Aswal VK, Hassan PA, Bahadur P. Influence of a hydrophobic diol on the micellar transitions of pluronic P85 in aqueous solution. J Colloid Interface Sci. 2008; 320(2):452-9. doi: 10.1016/j.jcis.2008.01.050, PMID 18275966.

  96. Lee CF, Yang CH, Lin TL, Bahadur P, Chen LJ. Role of molecular weight and hydrophobicity of amphiphilic tri-block copolymers in temperature-dependent co-micellization process and drug solubility. Colloids Surf B Biointerfaces. 2019;183:110461. doi: 10.1016/j.colsurfb.2019.110461.

  97. Wang Q, Li L, Jiang S. Effects of a PPO− PEO− PPO triblock copolymer on micellization and gelation of a PEO− PPO− PEO triblock copolymer in aqueous solution. Langmuir. 2005;21(20):9068-75. doi: 10.1021/la051537z, PMID 16171334.

  98. Sarkar B, Ravi V, Alexandridis P. Micellization of amphiphilic block copolymers in binary and ternary solvent mixtures. J Colloid Interface Sci. 2013;390(1):137-46. doi: 10.1016/j.jcis.2012.09.028, PMID 23099248.

  99. Lai TC, Kataoka K, Kwon GS. Pluronic-based cationic block copolymer for forming pDNA polyplexes with enhanced cellular uptake and improved transfection efficiency. Biomaterials. 2011;32(20):4594-603. doi: 10.1016/j.biomaterials.2011.02.065, PMID 21453964.

  100. Lu Y, Zhang X, Fan Z, Du B. Adsorption of PNIPAm110-PEO100-PPO65-PEO100-PNIPAm110 pentablock terpolymer on hydrophobic gold. Polymer. 2012; 53(17):3791-801. doi: 10.1016/j.polymer.2012.06.022. 

  101. Cho EB, Choi E, Yang S, Jaroniec M. Hollow mesoporous organosilica nanospheres templated with flower-like micelles of pentablock copolymers. J Colloid Interface Sci. 2018;528:124-34. doi: 10.1016/j.jcis.2018.05.076, PMID 29843060.

  102. Vyas B, Pillai SA, Tiwari S, Bahadur P. Effects of head group and counter-ion variation in cationic surfactants on the microstructures of EO-PO block copolymer micelles. Colloids Interface Sci Commun. 2019;33. doi: 10.1016/j.colcom.2019.100216, PMID 100216.

  103. Chen CY, Wang YC, Hung CC. In vitro dual-modality chemo-photodynamic therapy via stimuli-triggered polymeric micelles. React Funct Polym. 2016;98:56-64. doi: 10.1016/j.reactfunctpolym.2015.11.008.

  104. Gan H, Chen L, Sui X, Wu B, Zou S, Li A, Zhang Y, Liu X, Wang D, Cai S, Liu X, Liang Y, Tang X. Enhanced delivery of sorafenib with anti-GPC3 antibody-conjugated TPGS-b-PCL/Pluronic P123 polymeric nanoparticles for targeted therapy of hepatocellular carcinoma. Mater Sci Eng C Mater Biol Appl. 2018;91:395-403. doi: 10.1016/j.msec.2018.05.011, PMID 30033270.

  105. Hao J, Tong T, Jin K, Zhuang Q, Han T, Bi Y, Wang J, Wang X. Folic acid-functionalized drug delivery platform of resveratrol based on pluronic 127/D-α-tocopheryl polyethylene glycol 1000 succinate mixed micelles. Int J Nanomed. 2017;12:2279-92. doi: 10.2147/IJN.S130094, PMID 28392687.

  106. Xiong XY, Pan X, Tao L, Cheng F, Li ZL, Gong YC, Li YP. Enhanced effect of folated pluronic F87-PLA/TPGS mixed micelles on targeted delivery of paclitaxel. Int J Biol Macromol. 2017;103:1011-8. doi: 10.1016/j.ijbiomac.2017.05.136, PMID 28552723.

  107. Lübtow MM, Haider MS, Kirsch M, Klisch S, Luxenhofer R. Like dissolves like? A comprehensive evaluation of partial solubility parameters to predict polymer-drug compatibility in ultrahigh drug-loaded polymer micelles. Biomacromolecules. 2019;20(8):3041-56. doi: 10.1021/acs.biomac.9b00618, PMID 31318531.

  108. Xiao B, Zhang M, Viennois E, Zhang Y, Wei N, Baker MT, Jung Y, Merlin D. Inhibition of MDR1 gene expression and enhancing cellular uptake for effective colon cancer treatment using dual-surface-functionalized nanoparticles. Biomaterials. 2015;48:147-60. doi: 10.1016/j.biomaterials.2015.01.014, PMID 25701040.

  109. Zhang Y, Song W, Geng J, Chitgupi U, Unsal H, Federizon J, Rzayev J, Sukumaran DK, Alexandridis P, Lovell JF. Therapeutic surfactant-stripped frozen micelles. Nat Commun. 2016;7:11649. doi: 10.1038/ncomms11649, PMID 27193558.

  110. Kim BJ, Im SS, Oh SG. Investigation on the solubilization locus of aniline-HCl salt in SDS micelles with 1H NMR spectroscopy. Langmuir. 2001;17(2):565-6. doi: 10.1021/la0012889.

  111. Jackson JK, Springate CM, Hunter WL, Burt HM. Neutrophil activation by plasma opsonized polymeric microspheres: inhibitory effect of pluronic F127. Biomaterials. 2000;21(14):1483-91. doi: 10.1016/s0142-9612(00)00034-x, PMID 10872777.

  112. Lee ES, Oh YT, Youn YS, Nam M, Park B, Yun J, Kim JH, Song HT, Oh KT. Binary mixing of micelles using pluronics for a nano-sized drug delivery system. Colloids Surf B Biointerfaces. 2011;82(1):190-5. doi: 10.1016/j.colsurfb.2010.08.033, PMID 20850281.

  113. Tiwari S, Tirosh B, Rubinstein A. Increasing the affinity of cationized polyacrylamide-paclitaxel nanoparticles towards colon cancer cells by a surface recognition peptide. Int J Pharm. 2017;531(1):281-91. doi: 10.1016/j.ijpharm.2017.08.092, PMID 28844903.

  114. Luo YY, Xiong XY, Cheng F, Gong YC, Li ZL, Li YP. The targeting properties of folate-conjugated pluronic F127/poly (lactic-co-glycolic) nanoparticles. Int J Biol Macromol. 2017;105(1):711-9. doi: 10.1016/j.ijbiomac.2017.07.085, PMID 28716749.

  115. Song H, He R, Wang K, Ruan J, Bao C, Li N, Ji J, Cui D. Anti-HIF-1α antibody-conjugated pluronic triblock copolymers encapsulated with paclitaxel for tumor targeting therapy. Biomaterials. 2010;31(8):2302-12. doi: 10.1016/j.biomaterials.2009.11.067, PMID 20004970.

  116. Zhang W, Shi Y, Chen Y, Ye J, Sha X, Fang X. Multifunctional pluronic P123/F127 mixed polymeric micelles loaded with paclitaxel for the treatment of multidrug-resistant tumors. Biomaterials. 2011;32(11):2894-906. doi: 10.1016/j.biomaterials.2010.12.039, PMID 21256584.

  117. Minko T, Batrakova EV, Li S, Li Y, Pakunlu RI, Alakhov VY, Kabanov AV. Pluronic block copolymers alter apoptotic signal transduction of doxorubicin in drug-resistant cancer cells. J Controlled Release. 2005;105(3):269-78. doi: 10.1016/j.jconrel.2005.03.019, PMID 15939500.

  118. Alakhova DY, Kabanov AV. Pluronics and MDR reversal: an update. Mol Pharm. 2014;11(8):2566-78. doi: 10.1021/mp500298q, PMID 24950236.

  119. Khaliq NU, Park DY, Yun BM, Yang DH, Jung YW, Seo JH, Hwang CS, Yuk SH. Pluronics: intelligent building units for targeted cancer therapy and molecular imaging. Int J Pharm. 2019;556:30-44. doi: 10.1016/j.ijpharm.2018.11.064, PMID 30529667.

  120. Li Z, Qiu L, Chen Q, Hao T, Qiao M, Zhao H, Zhang J, Hu H, Zhao X, Chen D, Mei L. pH-sensitive nanoparticles of poly (l-histidine)–poly (lactide-co-glycolide)–tocopheryl polyethylene glycol succinate for anti-tumor drug delivery. Acta Biomater. 2015;11:137-50. doi: 10.1016/j.actbio.2014.09.014, PMID 25242647.

  121. Wu W, Wang J, Lin Z, Li X, Li J. Tumor‐acidity activated surface charge‐conversion of polymeric nanocarriers for enhanced cell adhesion and targeted drug release. Macromol Rapid Commun. 2014;35(19):1679-84. doi: 10.1002/marc.201400362, PMID 25171076.

  122. Lee ES, Gao Z, Bae YH. Recent progress in tumor pH targeting nanotechnology. J Controlled Release. 2008;132(3):164-70. doi: 10.1016/j.jconrel.2008.05.003, PMID 18571265.

  123. Liang Y, Su Z, Yao Y, Zhang N. Preparation of pH-sensitive pluronic-docetaxel conjugate micelles to balance the stability and controlled release issues. Materials (Basel). 2015;8(2):379-91. doi: 10.3390/ma8020379, PMID 28787944.

  124. Liu J, Huang Y, Kumar A, Tan A, Jin S, Mozhi A, Liang XJ. pH-sensitive nano-systems for drug delivery in cancer therapy. Biotechnol Adv. 2014;32(4):693-710. doi: 10.1016/j.biotechadv.2013.11.009, PMID 24309541.

  125. Xu C, Xu J, Zheng Y, Fang Q, Lv X, Wang X, Tang R. Active-targeting and acid-sensitive pluronic prodrug micelles for efficiently overcoming MDR in breast cancer. J Mater Chem B. 2020;8(13):2726-37. doi: 10.1039/c9tb02328c, PMID 32154530.

  126. Golwala P, Rathod S, Patil R, Joshi A, Ray D, Aswal VK, Bahadur P, Tiwari S. Effect of cosurfactant addition on phase behavior and microstructure of a water-dilutable microemulsion. Colloids Surf B Biointerfaces. 2020;186:110736. doi: 10.1016/j.colsurfb.2019.110736.

  127. Cheng X, Zeng X, Zheng Y, Fang Q, Wang X, Wang J, Tang R. pH-sensitive pluronic micelles combined with oxidative stress amplification for enhancing multidrug resistance breast cancer therapy. J Colloid Interface Sci. 2020;565:254-69. doi: 10.1016/j.jcis.2020.01.029, PMID 31978788.

  128. Wang H, Jiang H, Corbet C, de Mey S, Law K, Gevaert T, Feron O, De Ridder M. Piperlongumine increases the sensitivity of colorectal cancer cells to radiation: involvement of ROS production via dual inhibition of glutathione and thioredoxin systems. Cancer Lett. 2019;450:42-52. doi: 10.1016/j.canlet.2019.02.034, PMID 30790679.

  129. Deepagan VG, Kwon S, You DG, Nguyen VQ, Um W, Ko H, Lee H, Jo DG, Kang YM, Park JH. In situ diselenide-crosslinked polymeric micelles for ROS-mediated anticancer drug delivery. Biomaterials. 2016;103:56-66. doi: 10.1016/j.biomaterials.2016.06.044, PMID 27372421.

  130. Ji S, Xia J, Xu H. Dynamic chemistry of selenium: Se–N and Se–Se dynamic covalent bonds in polymeric systems. ACS Publications; 2016.

  131. Tong R, Lu X, Xia H. A facile mechanophore functionalization of an amphiphilic block copolymer towards remote ultrasound and redox dual stimulus responsiveness. Chem Commun (Camb). 2014;50(27):3575-8. doi: 10.1039/c4cc00103f, PMID 24566678.

  132. Li F, Xie C, Cheng Z, Xia H. Ultrasound responsive block copolymer micelle of poly (ethylene glycol)–poly (propylene glycol) obtained through click reaction. Ultrason Sonochem. 2016;30:9-17. doi: 10.1016/j.ultsonch.2015.11.023, PMID 26703197.

  133. Huang A, Zheng H, Wu Z, Chen M, Huang Y. Circular RNA-protein interactions: functions, mechanisms, and identification. Theranostics. 2020;10(8):3503-17. doi: 10.7150/thno.42174, PMID 32206104.

  134. Luo L, Zhang Q, Luo Y, He Z, Tian X, Battaglia G. Thermosensitive nanocomposite gel for intra-tumoral two-photon photodynamic therapy. J Controlled Release. 2019;298:99-109. doi: 10.1016/j.jconrel.2019.01.019, PMID 30703391.

  135. Mukerji R, Schaal J, Li X, Bhattacharyya J, Asai D, Zalutsky MR, Chilkoti A, Liu W. Spatiotemporally photoradiation-controlled intratumoral depot for the combination of brachytherapy and photodynamic therapy for solid tumor. Biomaterials. 2016;79:79-87. doi: 10.1016/j.biomaterials.2015.11.064, PMID 26702586.

  136. Shi S, Zhang L, Zhu M, Wan G, Li C, Zhang J, Wang Y, Wang Y. Reactive oxygen species-responsive nanoparticles based on peglated prodrug for targeted treatment of oral tongue squamous cell carcinoma by combining photodynamic therapy and chemotherapy. ACS Appl Mater Interfaces. 2018;10(35):29260-72. doi: 10.1021/acsami.8b08269, PMID 30106279.

  137. Uthaman S, Pillarisetti S, Mathew AP, Kim Y, Bae WK, Huh KM, Park IK. Long circulating photoactivable nanomicelles with tumor localized activation and ROS triggered self-accelerating drug release for enhanced locoregional chemo-photodynamic therapy. Biomaterials. 2020;232:119702. doi: 10.1016/j.biomaterials.2019.119702.

  138. Torchilin VP. Structure and design of polymeric surfactant-based drug delivery systems. J Controlled Release. 2001;73(2-3):137-72. doi: 10.1016/s0168-3659(01)00299-1, PMID 11516494.

  139. Wang Y, Yu L, Han L, Sha X, Fang X. Difunctional pluronic copolymer micelles for paclitaxel delivery: synergistic effect of folate-mediated targeting and pluronic-mediated overcoming multidrug resistance in tumor cell lines. Int J Pharm. 2007;337(1-2):63-73. doi: 10.1016/j.ijpharm.2006.12.033, PMID 17289311.

  140. Zhang W, Shi Y, Chen Y, Yu S, Hao J, Luo J, Sha X, Fang X. Enhanced antitumor efficacy by paclitaxel-loaded pluronic P123/F127 mixed micelles against non-small cell lung cancer based on passive tumor targeting and modulation of drug resistance. Eur J Pharm Biopharm. 2010;75(3):341-53. doi: 10.1016/j.ejpb.2010.04.017, PMID 20451605.

  141. Jain TK, Richey J, Strand M, Leslie-Pelecky DL, Flask CA, Labhasetwar V. Magnetic nanoparticles with dual functional properties: drug delivery and magnetic resonance imaging. Biomaterials. 2008;29(29):4012-21. doi: 10.1016/ j.biomaterials.2008.07.004, PMID 18649936.

  142. Jain TK, Morales MA, Sahoo SK, Leslie-Pelecky DL, Labhasetwar V. Iron oxide nanoparticles for sustained delivery of anticancer agents. Mol Pharm. 2005;2(3):194-205. doi: 10.1021/ mp0500014, PMID 15934780.

  143. Liu TY, Hu SH, Liu KH, Shaiu RS, Liu DM, Chen SY. Instantaneous drug delivery of magnetic/thermally sensitive nanospheres by a high-frequency magnetic field. Langmuir. 2008;24(23):13306-11. doi: 10.1021/la801451v, PMID 18954093.

  144. Bae WK, Park MS, Lee JH, Hwang JE, Shim HJ, Cho SH, Kim DE, Ko HM, Cho CS, Park IK, Chung IJ. Docetaxel-loaded thermoresponsive conjugated linoleic acid-incorporated poloxamer hydrogel for the suppression of peritoneal metastasis of gastric cancer. Biomaterials. 2013;34(4):1433-41. doi: 10.1016/j.biomaterials.2012.10.077, PMID 23174142.

  145. Li YP, Sun LZ, Xiong XY, Li ZL, Xing TK, Yao LH. Controlled Release characteristics of PLA-pluronic-PLA nano-sized vesicles in vitro. In: advanced materials research. AMR. 2013;785-786:493-7. doi: 10.4028/www.scientific.net/AMR.785-786.493.

  146. Lin H-R, Li Y-S, Lin Y-J. Novel microencapsulated pluronic–chitosan nanomicelles for lung delivery. Colloid Polym Sci. 2016;294(7):1209-16. doi: 10.1007/s00396-016-3879-6.

  147. Zhang J, Li Y, Fang X, Zhou D, Wang Y, Chen M. TPGS-g-PLGA/Pluronic F68 mixed micelles for tanshinone IIA delivery in cancer therapy. Int J Pharm. 2014;476(1-2):185-98. doi: 10.1016/j.ijpharm.2014.09.017, PMID 25223472.

  148. Adams ML, Lavasanifar A, Kwon GS. Amphiphilic block copolymers for drug delivery. J Pharm Sci. 2003;972(7):1343-55. doi: 10.1002/jps.10397, PMID 12820139.

  149. Bae YH, Yin H. Stability issues of polymeric micelles. J Controlled Release. 2008;131(1):2-4. doi: 10.1016/j.jconrel.2008.06.015, PMID 18625275.

  150. Barratt G. Colloidal drug carriers: achievements and perspectives. Cell Mol Life Sci CMLS. 2003; 60(1):21-37. doi: 10.1007/s000180300002, PMID 12613656.

  151. Li Y, Kwon GS. Methotrexate esters of poly (ethylene oxide)-block-poly (2-hydroxyethyl-L-aspartamide). Part I: Effects of the level of methotrexate conjugation on the stability of micelles and on drug release. Pharm Res. 2000;17(5):607-11. doi: 10.1023/a:1007529218802, PMID 10888314.

  152. Aliabadi HM, Shahin M, Brocks DR, Lavasanifar A. Disposition of drugs in block copolymer micelle delivery systems: from discovery to recovery. Clin Pharmacokinet. 2008;47(10):619-34. doi: 10.2165/00003088-200847100-00001, PMID 18783294.

  153. Oh KS, Song JY, Cho SH, Lee BS, Kim SY, Kim K, Jeon H, Kwon IC, Yuk SH. Paclitaxel-loaded Ppluronic nanoparticles formed by a temperature-induced phase transition for cancer therapy. J Controlled Release. 2010;148(3):344-50. doi: 10.1016/ j.jconrel.2010.08.021, PMID 20797418.

  154. Lee KE, Kim BK, Yuk SH. Biodegradable polymeric nanospheres formed by the temperature-induced phase transition in a mixture of poly (lactide-co-glycolide) and poly (ethylene oxide)− poly (propylene oxide)− poly (ethylene oxide) triblock copolymer. Biomacromolecules. 2002;3(5):1115-9. doi: 10.1021/bm020066h, PMID 12217061.

  155. Gautier S, Grudzielski N, Goffinet G, De Hassonville SH, Delattre L, Jerojme R. Preparation of poly (D, L-lactide) nanoparticles assisted by amphiphilic poly (methyl methacrylate-co-methacrylic acid) copolymers. J Biomater Sci Polym Ed. 2001; 12(4):429-50. doi: 10.1163/156856201750195306, PMID 11436978.

  156. Yuk SH, Oh KS, Koo H, Jeon H, Kim K, Kwon IC. Multi-core vesicle nanoparticles based on vesicle fusion for delivery of chemotherapic drugs. Biomaterials. 2011;32(31):7924-31. doi: 10.1016/j.biomaterials.2011.07.017, PMID 21784512.

  157. Firestone MA, Wolf AC, Seifert S. Small-angle X-ray scattering study of the interaction of poly (ethylene oxide)-b-poly (propylene oxide)-b-poly (ethylene oxide) triblock copolymers with lipid bilayers. Biomacromolecules. 2003;4(6):1539-49. doi: 10.1021/bm034134r, PMID 14606878.

  158. Oh KS, Kim K, Yoon BD, Lee HJ, Park DY, Kim EY, et al, Lee K, Seo JH, Yuk SH. Docetaxel-loaded multilayer nanoparticles with nanodroplets for cancer therapy. Int J Nanomedicine. 2016;11:1077-87. doi: 10.2147/IJN.S100170, PMID 27042062.

  159. Lu RM, Chang YL, Chen MS, Wu HC. Single chain anti-c-Met antibody conjugated nanoparticles for in vivo tumor-targeted imaging and drug delivery. Biomaterials. 2011;32(12):3265-74. doi: 10.1016/j.biomaterials.2010.12.061, PMID 21306768.

  160. Peer D, Karp JM, Hong S, Farokhzad OC, Margalit R, Langer R. Nanocarriers as an emerging platform for cancer therapy. Nat Nanotechnol. 2007;2(12):751-60. doi: 10.1038/nnano.2007.387, PMID 18654426.

  161. Gelperina S, Kisich K, Iseman MD, Heifets L. The potential advantages of nanoparticle drug delivery systems in chemotherapy of tuberculosis. Am J Respir Crit Care Med. 2005;172(12):1487-90. doi: 10.1164/rccm.200504-613PP, PMID 16151040.

  162. Haseeb MT, Khaliq NU, Yuk SH, Hussain MA, Bashir S. Linseed polysaccharides based nanoparticles for controlled delivery of docetaxel: design, in vitro drug release and cellular uptake. J Drug Delivery Sci Technol. 2019;49:143-51. doi: 10.1016/j.jddst.2018.11.009.

  163. Park JH, Saravanakumar G, Kim K, Kwon IC. Targeted delivery of low molecular drugs using chitosan and its derivatives. Adv Drug Deliv Rev. 2010;62(1):28-41. doi: 10.1016/j.addr.2009.10.003, PMID 19874862.

  164. Kozlov MY, Melik-Nubarov NS, Batrakova EV, Kabanov AV. Relationship between pluronic block copolymer structure, critical micellization concentration and partitioning coefficients of low molecular mass solutes. Macromolecules. 2000;33(9):3305-13. doi: 10.1021/ma991634x.

  165. Alakhova DY, Zhao Y, Li S, Kabanov AV. Effect of doxorubicin/pluronic SP1049C on tumorigenicity, aggressiveness, DNA methylation and stem cell markers in murine leukemia. PloLOS OneNE. 2013;8(8):e72238. doi: 10.1371/journal.pone.0072238, PMID 23977261.

  166. Batrakova EV, Kabanov AV. Pluronic block copolymers: evolution of drug delivery concept from inert nanocarriers to biological response modifiers. J Controlled Release. 2008;130(2):98-106. doi: 10.1016/j.jconrel.2008.04.013, PMID 18534704.

  167. Valle JW, Lawrance J, Brewer J, Clayton A, Corrie P, Alakhov V, Ranson M. A phase II, window study of SP1049C as first-line therapy in inoperable metastatic adenocarcinoma of the oesophagus. J Clin Oncol. 2004;22(14_suppl):4195. doi: 10.1200/jco.2004.22.90140.4195.

  168. Pitto Barry A, Barry NPE. Pluronic® block-copolymers in medicine: from chemical and biological versatility to rationalization and clinical advances. Polym Chem. 2014;5(10):3291-7. doi: 10.1039/C4PY00039K.

  169. Maeda H. Toward a full understanding of the EPR effect in primary and metastatic tumors as well as issues related to its heterogeneity. Adv Drug Deliv Rev. 2015;91:3-6. doi: 10.1016/j.addr.2015.01.002, PMID 25579058.

  170. Oh KS, Lee H, Kim JY, Koo EJ, Lee EH, Park JH, et al, Kim SY, Kim K, Kwon IC, Yuk SH. The multilayer nanoparticles formed by layer by layer approach for cancer-targeting therapy. J Controlled Release. 2013;165(1):9-15. doi: 10.1016/j.jconrel.2012.10.013, PMID 23103984.

  171. Valle JW, Armstrong A, Newman C, Alakhov V, Pietrzynski G, Brewer J, Campbell S, Corrie P, Rowinsky EK, Ranson M. A phase 2 study of SP1049C, doxorubicin in P-glycoprotein-targeting pluronics, in patients with advanced adenocarcinoma of the esophagus and gastroesophageal junction. Invest New Drugs. 2011;29(5):1029-37. doi: 10.1007/s10637-010-9399-1, PMID 20179989.

  172. Krylova OO, Melik‐Nubarov NS, Badun GA, Ksenofontov AL, Menger FM, Yaroslavov AA. Pluronic L61 accelerates flip–flop and transbilayer doxorubicin permeation. Chem Eur J.istry. 2003;9(16):3930-6. doi: 10.1002/chem.200204621, PMID 12916119.

  173. Batrakova EV, Li S, Brynskikh AM, Sharma AK, Li Y, Boska M, Gong N, Mosley RL, Alakhov VY, Gendelman HE, Kabanov AV. Effects of pluronic and doxorubicin on drug uptake, cellular metabolism, apoptosis and tumor inhibition in animal models of MDR cancers. J Controlled Release. 2010;143(3):290-301. doi: 10.1016/j.jconrel.2010.01.004, PMID 20074598.

  174. Sun H, Meng Q, Tang S, Su J, Yin Q, Chen L, Gu W, Yu H, Zhang Z, Wang S, Li Y. Inhibition of bbreast ccancer mmetastasis by ppluronic ccopolymers with mmoderate hhydrophilic–llipophilic bbalance. Mol Pharm. 2015;12(9):3323-31. doi: 10.1021/acs.molpharmaceut.5b00319, PMID 26220770.

  175. Zhu P, Zhao N, Sheng D, Hou J, Hao C, Yang X, Zhu B, Zhang S, Han Z, Wei L, Zhang L. Inhibition of growth and metastasis of colon cancer by delivering 5-fluorouracil-loaded pluronic p85 copolymer micelles. Sci Rep. 2016;6:20896. doi: 10.1038/srep20896. PMID 26864651.

  176. Cai Y, Sun Z, Fang X, Fang X, Xiao F, Wang Y, Chen M. Synthesis, characterization and anticancer activity of pluronic F68–curcumin conjugate micelles. Drug Delivery. 2016;23(7):2587-95. doi: 10.3109/10717544.2015.1037970, PMID 26066393.

  177. Nguyen DH, Bae JW, Choi JH, Lee JS, Park KD. Bioreducible cross-linked Ppluronic micelles: pH-triggered release of doxorubicin and folate-mediated cellular uptake. J Bioact Compat Polym. 2013;28(4):341-54. doi: 10.1177/0883911513491642.

  178. Xiong XY, Tao L, Qin X, Li ZL, Gong YC, Li YP, Yang YJ, Liu ZY. Novel folated Ppluronic/poly (lactic acid) nanoparticles for targeted delivery of paclitaxel. RSC Adv. 2016;6(58):52729-38. doi: 10.1039/C6RA09271C.

  179. Kim JY, Choi WI, Kim YH, Tae G. Brain-targeted delivery of protein using chitosan- and RVG peptide-conjugated, pluronic-based nano-carrier. Biomaterials. 2013;34(4):1170-8. doi: 10.1016/j.biomaterials.2012.09.047, PMID 23122677.

  180. Huang Y, Liu W, Gao F, Fang X, Chen Y. C (RGDyK)-decorated pluronic micelles for enhanced doxorubicin and paclitaxel delivery to brain glioma. Int J Nanomedicine. 2016;11:1629-41. doi: 10.2147/IJN.S104162, PMID 27143884.

  181. Domingues C, Alvarez Lorenzo C, Concheiro A, Veiga F, Figueiras A. Nanotheranostic pluronic-like polymeric micelles: shedding light into the dark shadows of tumors. Mol Pharm. 2019;16(12):4757-74. doi: 10.1021/acs.molpharmaceut.9b00945, PMID 31633939.

  182. Jo Y, Choi N, Kim K, Koo HJ, Choi J, Kim HN. Chemoresistance of cancer cells: requirements of tumor microenvironment-mimicking in vitro models in anti-cancer drug development. Theranostics. 2018;8(19):5259-75. doi: 10.7150/thno.29098, PMID 30555545.

  183. Wang M, Zhao J, Zhang L, Wei F, Lian Y, Wu Y, Gong Z, Zhang S, Zhou J, Cao K, Li X, Xiong W, Li G, Zeng Z, Guo C. Role of tumor microenvironment in tumorigenesis. J Cancer. 2017;8(5):761-73. doi: 10.7150/jca.17648, PMID 28382138.

  184. Domingues CSDC, Serambeque BP, Laranjo Cândido MS, Marto CMM, Veiga FJB, Sarmento Antunes Cruz Ribeiro AB, Figueiras ARR, Botelho MFR, Dourado MARF AB. Epithelial‐mesenchymal transition and microRNAs: challenges and future perspectives in oral cancer. Head Neck. 2018;40(10):2304-13. doi: 10.1002/hed.25381, PMID 30120853.

  185. Zhu X, Anquillare EL, Farokhzad OC, Shi J. Polymer- and protein-based nanotechnologies for cancer theranostics. In: Cancer theranostics. Elsevier; 2014. p. 419-36.

  186. Jo SD, Ku SH, Won YY, Kim SH, Kwon IC. Targeted nanotheranostics for future personalized medicine: recent progress in cancer therapy. Theranostics. 2016;6(9):1362-77. doi: 10.7150/thno.15335, PMID 27375785.

  187. Ma Y, Huang J, Song S, Chen H, Zhang Z. Cancer‐targeted nanotheranostics: recent advances and perspectives. Small. 2016;12(36):4936-54. doi: 10.1002/smll.201600635, PMID 27150247.

  188. Giacomelli FC, Stepanek P, Schmidt V, Jager E, Jager A, Giacomelli C. Light scattering evidence of selective protein fouling on biocompatible block copolymer micelles. Nanoscale. 2012;4(15):4504-14. doi: 10.1039/c2nr30623a, PMID 22688571.

  189. Oh KS, Han H, Yoon BD, Lee M, Kim H, Seo DW, Seo JH, Kim K, Kwon IC, Yuk SH. Effect of HIFU treatment on tumor-targeting efficacy of docetaxel-loaded Ppluronic nanoparticles. Colloids Surf B Biointerfaces. 2014;119:137-44. doi: 10.1016/j.colsurfb.2014.05.007, PMID 24881526.

  190. Zhang Y, Feng L, Wang J, Tao D, Liang C, Cheng L, Hao E, Liu Z. Surfactant‐stripped micelles of near-infrared dye and paclitaxel for photoacoustic Iimaging guided photothermal‐chemotherapy. Small. 2018;14(44):e1802991. doi: 10.1002/smll.201802991:, 1802991PMID 30286285.

  191. Nagy-Simon T, Potara M, Craciun AM, Licarete E, Astilean S. IR780-dye loaded gold nanoparticles as new near-infrared activatable nanotheranostic agents for simultaneous photodynamic and photothermal therapy and intracellular tracking by surface-enhanced resonant Raman scattering imaging. J Colloid Interface Sci. 2018;517:239-50. doi: 10.1016/j.jcis.2018.02.007, PMID 29428811.

  192. Pellosi DS, Calori IR, de Paula LB, Hioka N, Quaglia F, Tedesco AC. Multifunctional theranostic pluronic mixed micelles improve targeted photoactivity of verteporfin in cancer cells. Mater Sci Eng C Mater Biol Appl. 2017;71:1-9. doi: 10.1016/j.msec.2016.09.064, PMID 27987651.

  193. Sokolov IL, Cherkasov VR, Tregubov AA, Buiucli SR, Nikitin MP. Smart materials on the way to theranostic nanorobots: molecular machines and nanomotors, advanced biosensors, and intelligent vehicles for drug delivery. Biochim Biophys Acta Gen Subj. 2017;1861(6):1530-44. doi: 10.1016/j.bbagen.2017.01.027, PMID 28130158.

  194. Oba M. Study on development of polymeric micellar gene carrier and evaluation of its functionality. Biol Pharm Bull. 2013;36(7):1045-51. doi: 10.1248/bpb.b13-00287, PMID 23811553.

  195. Varela Moreira A, Shi Y, Fens MHAM, Lammers T, Hennink WE, Schiffelers RM. Clinical application of polymeric micelles for the treatment of cancer. Mater Chem Front. 2017;1(8):1485-501. doi: 10.1039/C6QM00289G.

  196. Hua S, De Matos MBC, Metselaar JM, Storm G. Current trends and challenges in the clinical translation of nanoparticulate nanomedicines: pathways for translational development and commercialization. Front Pharmacol. 2018;9:790. doi: 10.3389/fphar.2018.00790, PMID 30065653.

  197. Wong JKL, Mohseni R, Hamidieh AA, MacLaren RE, Habib N, Seifalian AM. Limitations in clinical translation of nanoparticle-based gene therapy. Trends Biotechnol. 2017;35(12):1124-5. doi: 10.1016/j.tibtech.2017.07.009, PMID 28822599.

  198. Kadian R. Nanoparticles: a promising drug delivery approach. Asian J Pharm Clin Res. 2018;11(1):30-5. doi: 10.22159/ajpcr.2017.v11i1.22035.