• Open Access
    Review

    Nanoparticle-based targeted therapy through EGFR tyrosine kinase inhibitors and their recent advances in lung cancer therapy

    Priyanka S. Waghmare
    Anuruddha R. Chabukswar *
    Kunal G. Raut
    Bhagyashree Gaikwad-Pawar
    Swati C. Jagdale

    Explor Med. 2024;5:513–529 DOI: https://doi.org/10.37349/emed.2024.00236

    Received: January 31, 2024 Accepted: March 27, 2024 Published: July 14, 2024

    Academic Editor: Jingchao Li, Donghua University, China

    Abstract

    The emergence and subsequent advancement of nanotechnology in recent years have greatly benefited the healthcare sector, particularly in the treatment of cancer. As per study, major fatalities are related to the lung cancer. For many years, oral tyrosine kinase inhibitors (TKIs) against the epidermal growth factor receptor (EGFR) family of receptors have been used in the clinic to treat human malignancies, although they observed some very serious adverse effects in the treatment of lung cancer, especially in non-small cell lung cancer (NSCLC). Despite EGFR-TKIs’ exceptional qualities as small-molecule targeted medications, their applicability is nevertheless limited by their poor solubility, inconsistent oral bioavailability, high daily dose needs, high plasma albumin binding propensity, and initial/acquired drug resistance. Article’s purpose is to investigate EGFR-TKI’s effects on lung cancer and get around some of its drawbacks, nanotechnology will be an innovative strategy. An effective tool to increase the effectiveness of these pharmaceuticals is nanotechnology by methods other than oral. This article signifies that a range of nanomedicine delivery systems have been developed to effectively distribute EGFR-TKIs with improved drug release kinetics and tissue-targeting capacity. This review article intends to present information regarding lung cancer and EGFR relation, mechanism of recently approved EGFR-TKI’s targeted therapy, an updated landscape of EGFR-TKIs and their clinical status over lung cancer, advantages and disadvantages of nanotechnology, and new breakthroughs in nano-delivery which mentioned as a significantly better over traditional drug chemotherapy and delivery.

    Keywords

    Lung cancer, EGFR, tyrosine kinase inhibitor, nanotechnology

    Introduction

    A great deal of progress has been made in cancer-related treatments during the last few years, as cancer is often regarded as the greatest threat to people’s health and lives. Lung carcinoma is the most prevalent cancer and has a high incidence of cancer-related fatalities including both men and women globally [1, 2]. That’s to a 2018 analysis, lung carcinoma is the main cause of mortality related to cancer, accounting for 11.6 proportions of cancer diagnoses and 18.4 proportions of fatalities [1, 3]. Patients who are diagnosed in the proximal (60%), regionally (33%), or distance (6%) stages have significantly different 5-year relative survival rates [4]. Small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC) are indeed the two types of lung carcinoma, with NSCLC account for roughly 80% of cases and SCLC for 20% [5].

    The epidermal growth factor receptor is a glycoprotein with a transmembrane structure obtaining an external epidermal growth factor binding and an intracellular tyrosine kinase domain which is mainly expressed in some epithelial, mesenchymal and neurogenic tissue. It regulates signaling processes to govern the growth of cells by triggering many signal transduction cascade. This continuous process leads to the activation of tumor phenotype. Not only this sustained process but mutation in EGFR leads to malignancy in lungs like adenocarcinoma and sometime patients does not have survival benefit [6]. Traditional lung cancer chemotherapy treatments lack targeting tumor cells specifically, causing severe harm to general cells such as bone marrow arrest, gastrointestinal (GI) responses, and phlebophlogosis [7]. NSCLC generally responds to chemotherapy and radiotherapy less favorably than SCLC. In patients with advanced metastatic illness, chemotherapy-targeted medications, and other supportive therapies may boost survival and relieve symptoms [8].

    The discovery of lung cancer mutations has resulted in the development of biochemically tailored therapies to enable patients with metastatic conditions to live longer lives [9]. In most cases, the first-line therapy for NSCLC involves platinum-based chemotherapy, such as cisplatin or carboplatin doublets. However, it has very major negative effects [10]. NSCLC is characterized by multiple gene point mutations, with somatic mutations in the exons of the EGFR gene occurring in roughly 70% of NSCLC patients [11].

    Nanoparticles (NPs) are small artificial particles with a diameter of fewer than 100 nanometers made of polymers, lipids, or metals like gold. NPs are particularly useful in a variety of medicinal applications, ranging from diagnosis to cancer treatment [12]. These NPs are strikingly close in size to most biological structures and compounds. As a result, they provide functional features for cancer research both in vivo and in vitro [13].

    Role of EGFR-TKI in targeted therapy

    Patients with unique activating genetic variants in the EGFR tyrosine kinases region profit more from FDA-approved drugs (exons 18–21). The most frequent EGFR-activating alterations are exon 19 deletions (del19) (45 percent) and the L858R exon 21 replacement (40–45 percent). Exon 18 alteration make up just approximately 5 percent of all EGFR mutations, which is a significantly lower frequency [14, 15]. Figure 1 mentioned recently approved EGFR-TKI drugs as its targeted therapy for lung cancer.

    Functioning of tyrosine kinase receptor with recently approved inhibitory drugs [16]

    Note. Adapted from “The emerging treatment landscape of targeted therapy in non-small-cell lung cancer” by Yuan M, Huang LL, Chen JH, Wu J, Xu Q. Signal Transduct Target Ther. 2019;4:61 (https://www.nature.com/articles/s41392-019-0099-9). CC BY.

    Role of adjuvant EGFR-TKI

    Exon 19 deletion mutations, advanced disease stages, prolonged treatment, and the usage of third-generation EGFR-TKIs were all linked to greater disease-free survival (DFS) benefits. Combining therapy EGFR-TKIs should enhance DFS but not Overall Survival (OS) in EGFR mutation-positive patients with stage IB-IIIA NSCLC therapy EGFR-TKIs should enhance DFS but not OS in EGFR mutation-positive patients with stage IB-IIIA NSCLC when compared to subsequent chemotherapy or placebo. The illness stage, length of the treatment, types of mutation, and therapeutic drugs may all have an impact on the degree of effect. Adjuvant EGFR-TKIs demonstrated better tolerance than chemotherapy, especially when first-generation medicines were used [17]. Various clinical trials are undergoing like Wu 2020 (ADURA) [18], Zhong 2018 (ADJUVANT) [19], Yue 2018 (EVAN) [20], Li 2014 [21], Kelly 2015 (RADIANT) [22]. Numerous recent trials have looked into the efficacy of EGFR-TKI therapy as a preoperative treatment for NSCLC. Preoperative EGFR-TKI therapy has the capability to decrease tumor size, enhance radiological outcomes, and boost the likelihood of curative surgery. The effectiveness and safety of neoadjuvant EGFR-TKI treatment were assessed using a pooled analysis of prospectively trial information. Patients having treatable EGFR-variant NSCLC can get adjuvant chemotherapy, EGFR-TKI, or a combination of the two, albeit which is the most beneficial remains unknown. It should be looked into given the superior objective response rate, surgical resection rate, and safety of neoadjuvant EGFR-TKI over chemotherapy for potentially resectable NSCLC. Patients with operable EGFR-mutant NSCLC have access to subsequent chemotherapy, EGFR-TKI, or a combination of the two, while it is debatable which is the most effective. Given the improved objective response rate, surgical resection rate, and safety of preoperative EGFR-TKI over chemotherapy for potentially resectable NSCLC, it should be investigated [23]. Table 1 mentioned recently approved EGFR- TKI and their clinical status.

    Clinical Status of recently approved drugs [24, 25]. The fundamental functional principle and clinical aspect of the four FDA-approved EGFR-TKIs that are most frequently used are summarised in this gefitinib (Iressa), erlotinib (Tarceva), osimertinib (Tagrisso), afatinib (Gilotrif), and dacomitinib (Vizimpro) [26]

    TKIErlotinibGefitinibAfatinibDacomitinibOsimertinib
    Brand nameTarcevaIressaGilotrifVizimproTagrisso
    Dose150 mg 1 h before or 2 h after food250 mg +/− food40 mg 1 h before or 2 h after food45 mg orally once daily80 mg +/− food
    IndicationFirst generation for advanced NSCLCFirst generation for advanced NSCLCSecond generation for advanced NSCLCSecond generation for advanced NSCLCThird generation for advanced NSCLC
    Side effectsRash, diarrhea, edema, cough, conjunctivitisskin reaction, rash, anorexia, stomatitis diarrhea, paronychiaEruption rash dry skin, diarrhea, loss of appetite, stomatitis, diarrheaDiarrhea, interstitial lung disease, stomatitis, rashQTc prolongation, ocular disorder cardiomyopathy
    MOACompetitive; reversible
    EGFR L858R, Del19
    Competitive; reversible
    EGFR L858R, Del19
    Covalent; irreversible
    EGFR L858R, L858R/T790M
    Covalent; irreversible
    EGFR L858R, L858R/T790M
    Covalent; irreversible
    Drug approval year20042003201320122015
    StatusPhase III approved (2012)Phase III approved (2019)Phase III approved (2021)Phase III approved (2018)Phase III approved (2021)
    Display full size

    NSCLC: non-small cell lung cancer; MOA: mechanism of action

    Some recent combinations of EGFR-TKI

    In 2022, a novel family of EGFR-TKIs will be developed through structural changes of 2,4-dihydroindeno[1,2-c]pyrazoles. These chemicals were compared to erlotinib’s potency [27] (The sponsor decided to discontinue the trial after deciding to examine the pairing of quaratusugene ozeplasmid and osimertinib rather than quaratusugene ozeplasmid and erlotinib [28]). The US FDA granted fast-track designation to a pair of quaratusugene ozeplasmid immunogene therapy and pembrolizumab for the treatment of patients with relapsed stage III or IV NSCLC who had already worsened on pembrolizumab [29]. Currently, phase II and III trials are being conducted to compare osimertinib against osimertinib plus chemo treatment, such as cisplatin/carboplatin and pemetrexed. The OPAL Study (NEJ032C/LOGIK1801) is a phase II study of osimertinib in association with platinum with pemetrexed in patients suffering from advanced NSCLC who have EGFR mutations [30]. Combination chemotherapy can also boost the number of people in remission. The patient with EGFR-mutated advanced NSCLC underwent well-tolerated combination therapy and benefited from osimertinib in conjunction with chemotherapy and bevacizumab [31]. For first-line chemotherapy, a combination of osimertinib and gefitinib is acceptable [32] as well as second-line treatment (OSCILLATE) [33] of EGFR-modified NSCLC resulted in fast plasma clearance of the EGFR mutation. Dual EGFR inhibition with gefitinib + osimertinib may postpone the establishment of acquired resistance, according to preclinical research [32].

    Just for knowledge, osimertinib was compared to afatinib for the treating of 790M-positive NSCLC and numerous central nervous system (CNS) lesions after lack of first EGFR-TKI medication in an Asian population, and osimertinib was found to be superior to afatinib [34].

    Nanotechnology in cancer

    Regardless of the fact that chemotreatment and radiotherapy are treatment options for lung malignancy, the majority of patients experience a recurrence that is more resistant to subsequent treatment. A promising pharmacological approach is required to improve the prognosis of this type of cancer. Significant improvements in the research and the utilization of nanotechnology in the diagnostic and treatment of this form of cancer have been made in recent years, including the usage of LBNPs [35]. Nanomedicine is a comparatively recent treatment that aims to replace drug delivery and boost therapeutic advantages while limiting negative impacts on healthy tissues [36]. Blood circulation and clearance of the drug provide balance between its activity and toxicity. Due to the physicochemical properties of the nanomaterials, it mainly distributed in lung, liver, and kidney hence their clearance is expected to occur from these organs. The safety and effectiveness of nanocarriers are not assured and cannot be precisely managed due to the unknown processes of absorption, distribution, metabolism, and excretion, also the drug release mechanism of nanocarriers in the body. Therefore, they are rarely approved for clinical usage [37, 38]. On consideration of toxicity human body there is 2 types of toxicity e.g., primary [inflammation, oxidative stress, deoxyribonucleic acid (DNA) damage by direct exposure to organs like lung, skin, etc.] and secondary (inflammation, ROS generation, altered cellular function, etc.) [39]. Therefore, very least are actually there in clinical trials recently for lung cancer as mentioned in Table 2.

    Clinical trials of EGFR-TKI with nanoparticle in lung cancer. In August 2020, a clinical trials.gov advanced search for “nanoparticles and lung cancer” was conducted. These were evaluated and chosen based on the study’s current status.

    Sr. No.Clinical trialStudy typeDescriptionStatus
    1NCT00553462Phase IIThis phase II trial is studying how well giving carboplatin and paclitaxel albumin-stabilized nanoparticle formulation together with radiation therapy and erlotinib works in treating patients with stage III NSCLC that cannot be removed by surgery.Completed
    2NCT04486833Phase I–IISafety and efficacy of GPX-001 + osimertinib in NSCLC patients with activating EGFR mutations who have progressed while on treatment with osimertinib.Recruiting
    Display full size

    Despite the success of different drug delivery systems in recent years, there are yet certain challenges that be resolved and advanced technologies to be developed in order to successfully carry pharmaceuticals to their local sites. As a result, nano-based drug delivery devices are still researched to aid in improved drug delivery [40]. Two types of NPs used in the treatment of lung cancer, organic NPs (liposomes, solid lipid nanoparticle, nanostructued liquid carriers, polymeric NP, colloidal polymeric micelles, dendrimer, etc.) and inorganic NPs (quantum dots, magnetic NPs, carbon nanotubes, etc.) [41]. Figure 2 mentioned the various nanomaterials depending on their physicochemical properties.

    Describing physicochemical characteristics of nanoparticle. SLN: solid lipid nanoparticle; NLC: nanostructured lipid carriers; NP: nanoparticle

    There are numerous advantages of using NPs over other materials. Because of the enhanced permeability and retention (EPR) action, NPS can stay in the tumour location. Another virtue of NPS is that its surface may be easily modified to reduce inappropriate ingestion by the reticuloendothelial system [42]. The large surface-area-to-volume ratio is another feature that permits therapeutic compounds to be held successfully and not degraded by the microenvironment. Nanomaterials are also being used to produce more effective and regulated drug delivery techniques [40]. Nanotechniques have proven to bridge the gap among biological and morphological sciences through the use of nanostructures and nanophases in a wide range of scientific domains [43]. Such particles are very important in nanomaterials and medication delivery systems based on nanotechnology [44, 45].

    Advantages of NPs

    Nanostructures stay in the bloodstream for a long period of time, allowing coupled drugs to be delivered at the appropriate dose. As a result, they cause less plasma fluctuations and have fewer negative impacts. These nanostructures penetrate the tissue matrix, allowing for easy drug uptake by cells, efficient drug distribution, and targeted action. Nanostructures are substantially easier for cells to consume than large particles with sizes ranging from 1 to 10 µm [46, 47]. NPs provide the best way to deliver natural compounds with high safety and efficacy. These nanomaterials are labeled with drugs that have minimal penetration and ingestion [47, 48]. Thus, nanotechnology provides relevant qualifications in the treatment of lung cancer via location specific and goal-oriented medication delivery [40]. NP based medicines developed not just for treatment but also for diagnosis, imaging, detection, etc. of the lung cancer [49].

    Application of NPs

    Getting the drug to the proper place in the body while limiting any unwanted effects on healthy organs is one of the most complex aspects of drug transmission. This is especially challenging in cure for cancer because the tumor may appear as an organ lesion. These strategies are aided by certain NPs. These benefits include their tiny size, which enables them to pass through cell membranes, link and stabilize peptides, and exit from lysosomes following endocytic [50]. The goal of drug capture in nanotechnology is to either boost release of the drug to or ingestion by specific receptors, or to reduce the toxic effects of the free drug to non-target systems. Lengthy and target-specific nanomaterials are required to achieve these goals. NP capture in the liver and spleen’s cellular eosinophils system is one of the issues [51]. Irrespective of how the treatment is administered, the bloodstream’s bioavailability permits active substance to travel to and contact practically all tissues throughout the body, and once there is drug content in the circulation, it can pass through endothelium obstacles. To build exact localization, a transporter that can disperse the medicine directly towards the target area can be used [52]. After NPs reach the intended region via systemic circulation, the interaction of ligands and receptors is referred to as active targeting. Only close proximity (0.5 nm) between the two components allows for ligand-receptor interaction [53]. Polyethylene glycol (PEG) surface renovation culminated in longer bloodstream circulation by blocking identification and consumption by the single-nuclear engulfing system [54]. Surface charge and other chemical characteristics may influence NP fate in cells. Gold NPs with PEG surfaces were effectively up taken in endosomes and cytoplasm, with nuclear localization [55]. Cells ingested poly(DL-lactide-co-glycolide) NPs by endocytosis [56, 57]. carbohydrate-binding agents to increase cellular contact on the interface of both biocompatible and biodegradable polylactic-co-glycolic acid (PLGA) nanospheres [58]. When the drug is administered as NPs or incorporated into them, higher adherence may contribute to improved drug activity. The attachment of certain peptides, like antibodies, to the nanomaterial surface may allow for more precise immunologically directed particle localization [59, 60]. Antibodies (immunoglobulin) are a crucial component in the formation of a system that actively targets NPs. Single-chain segment factors and antibody antigen-binding segments have two lighter and heavier chains that can bind sensors or antigen, respectively [61]. Folate, often known as folic acid belongs to the B vitamin family. It is a glycosylphosphatidylinositol-anchored cell-surface receptor, and this system is a ligand that is ideal for targeting tumor cells [62]. Aptamers are non-toxic targeting ligands that have more flexibility, stability, low molecular weight, and repeatability, than natural antibodies [52]. Table 3 mentioned the recent NP formulations of EGFR TK anticancer drugs.

    Summary of the recently published studies about the EGFR TK anticancer drugs-loaded Nanoparticulate formulations for the treatment of LC

    Sr. No. DrugNanoparticle formulationAim/DescriptionYearRef
    1AfatinibLiposomeLPN modified with FD7 or CCD as a delivery platform was explored to enhance afatinib delivery across the BBB model of mouse brain-derived endothelial bEnd.3 cells.2021[63]
    2OsimertinibLiposome vesicleLiposome-based delivery of OSI can provide a new formulation of the drug that can be administered via alternative delivery routes (intravenous, inhalation).2020[64]
    3GefitinibLiposomeGefitinib encapsulation based on nano liposome for enhancing curative effect of lung cancer2020[65]
    4GefitinibSLNPreparation and evaluation of inhalable dry powder containing glucosamine-conjugated gefitinib SLNs for lung cancer therapy2020[66]
    5GefitinibLiposomeLiposomal gefitinib dry powder inhalers (LGDs) were prepared using the injection-lyophilization method2020[67]
    6AfatinibLuminescence nanoparticlesNext-generation cancer-specific hybrid theranostic nanomaterials: MAGE-A3 NIR persistent luminescence nanoparticles conjugated to afatinib for in situ suppression of lung adenocarcinoma growth and metastasis2020[68]
    8ErlotinibSLNCo-encapsulated nanoparticles of Erlotinib and Quercetin for targeting lung cancer through nuclear EGFR and PI3K/AKT inhibition2022[69]
    9ErlotinibMesoporous silica nanoparticleInjectable thermosensitive hydrogel containing erlotinib-loaded hollow mesoporous silica nanoparticles as a localized drug delivery system for NSCLC therapy2020[70]
    Display full size

    LPN: lipid polymeric nanoparticle; BBB: blood brain barrier; SLN: solid lipid nanoparticle; EGFR: epidermal growth factor receptor; NSCLC: non-small cell lung cancer

    Table 4 mentioned newer combinations of anticancer NP formulations respectively.

    Some newer combinations of TKI

    Sr. No.DrugNanoparticle formulationDescription YearRef
    1Icotinib & doxorubicinEDS nanoparticleIn conclusion, the present study demonstrated that the use of synergistic NPs was effective for in vitro and in vivo targeted delivery and the inhibition of NSCLC and greatly enhanced the synergistic effect of doxorubicin and icotinib2020[71]
    2Erlotinib & bevacizumabpH-sensitive lipid-polymer hybrid nanoparticlesNon-small cell lung cancer combination therapy: hyaluronic acid modified, epidermal growth factor receptor-targeted, pH-sensitive lipid-polymer hybrid nanoparticles for the delivery of erlotinib plus bevacizumab2020[72]
    3Erlotinib & bevacizumabMagnetic nanoparticleA multifunctional nanotheranostics agent potentiates erlotinib to EGFR wild-type non-small cell lung cancer2022[73]
    4Gefitinib & thymoquinonePLGA nanoparticleDelivery of gefitinib in synergism with thymoquinone via transferrin-conjugated nanoparticle sensitizes gefitinib-resistant non-small cell lung carcinoma to control metastasis and stemness2021[74]
    5Osimertinib & selumetinibMicelleNanoparticles for co-delivery of osimertinib and selumetinib to overcome osimertinib-acquired resistance in non-small cell lung cancer2021[75]
    6Erlotinib & doxorubicinRhamnolipid-coated W/O/W double emulsion nanoparticlesRL-NP-DOX-ERL provided efficient delivery of doxorubicin and ERL to tumor tissue and successful tumor therapy with a synergetic effect2021[76]
    Display full size

    NPs: nanoparticles; NSCLC: non-small cell lung cancer; EGFR: epidermal growth factor receptor; PLGA: polylactic-co-glycolic acid; RL-NP-DOX-ERL: rhamnolipid-coated double emulsion nanoparticles containing doxorubicin and erlotinib

    Recent advances in EGFR-TKI drug NPs in cancer

    Organic NPs

    Liposome

    Alec Bangham was the first to find them in 1960. Liposomes are a well-known medication delivery technology used to enhance medicine distribution. They are circular packets made up of phospholipids and immunosuppressant with diameters that vary from 50 to 450 nm [77]. They have also been proven to stabilise medicinal molecules, promote biodistribution, work with both water loving and water phobic medications are available, and they are both biocompatible and biodegradable. Four categories of liposomes: (1) Water nucleus was wrapped by a lipid bilayer that could produce anionic, cationic, or neutral lipids and phospholipids in traditional liposomes. Water loving or water phobic materials can be used to infill both the cell membrane and the aquatic area in this scenario. (2) PEGylated varieties: PEG is introduced into the liposome’s interface to achieve steric saturation. (3) Ligand-targeted type: ligands such as polysaccharides, antibodies and peptides are bonded to the liposome’s interface or the ends of already binded PEG sequence and (4) Theragnostic liposome: This form of liposome is hybrid of the previous 3 categories, containing a NP as well as a targeting, imaging, and therapeutic component [78, 79]. Afatinib was encapsulated in “liposomes” (LPs) to allow for prolonged blood circulation and increased tumor permeability and retention. Cetuximab was also created to attach to drug-loaded liposomes, resulting in the formation of “immuno-liposomes” with tumor-cell selection and therapeutic action [80]. The ammonium sulfate gradient approach can be used to make afatinib liposomes with a greater encapsulation efficiency and a smaller particle size [81]. The pH-sensitive liposome with afatinib is a novel targeted medication delivery system for cancer treatment. Additionally, the inhibitory effect on H-1975 and HCC-827 was showed by demonstrating that afatinib-loaded pH-sensitive liposome had high anticancer activity [82].

    Solid lipid nanoparticle (SLN)

    SLNs are a novel dispersed drug administration technique constituted of biological oily material which remained solid at ambient and internal temperatures [35]. SLN has the potential to transport both hydrophilic and lipophilic medicines, as well as a high level of stability, extended bloodstream persistence, and the ability to be created from biocompatible materials [83]. These particles range in size from 50 to 1,000 nm. Solid lipids used as a solid material for drug encoring possessing monosaccharides, fatty acids, disaccharides, triglycerides, and compound glyceride mixtures. This matrix is stabilized by a mixture of surfactants or polymers [35]. SLNs pair the characteristics of a fat-emulsion carrier, liposome carrier, and polymeric NP, building them ideal for localized drug administration [84]. The ability of SLN to transfer the commonly used cytostatic (5-fluorouracil) through A549 cells’ resistance [85]. Treatment of NSCLC with microparticles containing erlotinib-loaded SLN by formulating dry powder inhaler to increase cargo drug molecule in alveoli adenocarcinoma over oral dosage of erlotinib [86]. Cetyl alcohol, Compritol 888 ATO, bee wax, cholesterol butyrate, and emulsifying wax are the most often utilized solid lipids for making SLN [87]. As per Wioletta Zielińska [88] possibilities for using SLNs in combination with 5-fluorouracil to defeat NSCLC cell line A549’s drug resistance. Unfortunately, like other colloidal systems, the SLNs have low drug loading, which is restricted by the drug’s solubility in the melting lipid phase and its ejection during storage [89, 90]. Afatinib-loaded SLN and paclitaxel delivered via PLGA porous microspheres dry powders: a newer therapy for EGFR inhibitors of tyrosine kinase NSCLC resistant. Author showed that combination of the EGFR-TKI with other chemotherapeutic agent would be feasible to overcome resistant over resistance produced by sing EGFR-TKI [91].

    Nanostructured lipid carriers (NLC)

    NLC (nanostructured lipid carriers) are second-generation lipid-based NPs that were first published in the mid-1990s to address the drawbacks of SLNs [92]. NLC are formed up of solid and liquid lipids that have been partially crystallized and dispersed in an aqueous phase containing an emulsifier [91]. This loosely packed crystalline structure allows drug molecules to be entrapped, reducing drug leakage during storage and allowing regulated drug release [93]. The NLC system has a low drug loading capacity because of the crystalline structure of the lipids and gelation in the dispersed phase due to solid lipid polymorphism [94]. In 2019, researchers compared erlotinib-loaded NLC and liposomes vs. the A549 lung cancer cell line in vitro. Erlotinib-NLC were more effective against cancer than erlotinib-liposomes mentioned in Table 4 [67].

    Polymeric NPs

    Polymeric NPs have already been widely studied in the treatment for cancer [95]. Polymers with a variety of structures, both natural and manufactured, can be used. Since they are biocompatible and biodegradable, polypeptides, albumin, gelatin, and chitosan are among the most often utilized natural polymers. Among the synthetic polymers utilized are PEG, polylactic acid, PLGA, and polycaprolactone [96]. Their use of polymeric NP allows maximum packaging of hydrophobic medicines, resulting in longer circulation durations and more effective enforcement at the site of action, as well as an improvement in therapeutic and targeting agent efficacy [97]. The goal of Vaidya’s study [98] was to create a NP method to overcome erlotinib resistance in patients of NSCLC. Erlotinib-cyclodextrin complex was synthesized using cyclodextrin sulfobutyl ether, which was then loaded into the core of biodegradable PLGA NPs via repeated emulsion solvent evaporation. Cationic polymers are known for their cellular toxicity and enhanced clumping in lung capillaries because they are non-degradable and poor compliance. As a result, constant monitoring of cationic polymer-induced toxicity is essential, as they cause dyspnea when they combine with a biosurfactant [99].

    Polymeric micelles

    Micelles are amphiphilic block copolymer nanomaterials that self-collected in a watery solution that build a conceptual model [40]. Polymeric micelles have a restricted dispersion to avoid too fast elimination through kidney, allowing them to concentrate in carcinomal parts via the EPR effect. Their polymeric coating also minimizes unwanted interactions with bioactive molecules. Their inner space structure allows for drug assimilation, which enhances stability and bioavailability, these nanomaterials have a lot of potential for water hating material transportation [100, 101]. The biodegradable nature of this technique for drug delivery systems, which may be employed for both cancer and eye medication administration, is its main advantage [41]. There are many formulations of EGFR-TKI which were signified for HER2 overexpression, synergistic effect, and multidrug resistance [102].

    Dendrimer

    Regardless of how the medication is delivered, the bioavailability of the blood permits drug particles to diffuse and get into practically all tissues in the body that are affected, and once the medication is in circulation, it crosses capillary boundaries. A carrier that redistributes the medicine selectively toward the target tissue can be employed to accomplish correct targeting [103]. Drug solubility and drug targeting (active and/or passive targeting) are both affected by dendrimers. Dendrimers have been used to diagnose diseases, particularly malignancies; consequently, we will focus on their diagnostic uses and briefly explore their capacity to deliver anticancer medications here [104]. Dendrimers offer a wide range of applications owing to their functional and structural diversity. They can be used in a variety of applications, possessing gene and small interfering ribonucleic acid (siRNA) delivery, pharmacy, photodynamic therapy, biomedicine, oligonucleotide conjugation, immunology, and imaging [105].

    Inorganic NP

    Metal-based NP

    Metal-based NPs such as gold, silver, carbon nanotubes, and quantum dots have all been studied as potential drug delivery methods in the treatment of NSCLC [106]. Magnetic NPs are made up of a magnetic inner side and an active layer over it. Materials like gold, iron, cobalt, and nickel build up the magnetic core, which gives it its magnetic qualities. The upper cover prevents clustering and decreases the magnetic core’s contact with other substances [107]. Metal-based NPs have shown exponential expansion in research due to their acceptable biocompatibility and simplicity of size manipulation and surface modification. Their ability to block visible light makes them ideal for intracellular tracking [108]. As per Alex Yang-Hao Yu, collagen gold NPs could be used as a platform for siRNA delivery because of the positive findings. Treatment with collagen-gold-EGFRsi resulted in noticeable tumour control effects, with tumour weight reduced by 30% for lipofetamine carriers and 70% for C-Au carriers [109].

    Carbon nanotubes

    Carbon nanotubes are hydrophobic tubular structures constituted of carbon atoms having diameters vary from 4 nm to 100 mm that change dependence on the graphene molecule arrangement. Carbon nanotubes are inherently not soluble in organic solvents or aqueous solutions, and the toxicity they generate in body fluids is a critical issue that must be resolved [110]. Due to its unique physicochemical properties like low cytotoxicity, bioavailability, etc. has ability to gather into cancer cells without any phagocyte recognition, and hence they achieve high intracellular drug concentration [111]. Carbon nanotubes may enter cells and distribute chemicals into the cytoplasm utilizing “needle-like penetration.” These NPs have a vast surface area, which allows for multiple attachment sites for potential targeting ligands, as well as an interior cavity that can hold therapeutic or diagnostic substances [36]. The parallels between asbestos and carbon nanotubes have sparked widespread worry about potential dangers and are still under investigation. Remember that the asbestos disaster may have been avoided [112]. Chemical treatment increases biocompatibility lowers toxicity and converts them into water-soluble nano-carriers [113].

    Conclusions

    We have summarized targeted therapy for NSCLC, especially through EGFR-TKI. Numerous receptors are there but EGFR is more commonly responsible for the metastasis of cancer. EGFR-TKIs directly act on the cellular membrane and directly stop tumor growth. These inhibitors are used in various ways to overcome EGFR resistance and to become more potent. This review listed recent advances in EGFR-TKI like new generation drugs and adjuvant therapy. Despite having such good qualities all these have some limitations as these produce adverse effects sometimes very serious. NPs are introduced to overcome side effects and to provide targeted release of these drugs. This study showed that introducing nanotechnology comes with better drug delivery and fewer side effects over traditionally used EGFR-TKI therapy for lung cancer. Types of NPs are introduced some are under clinical trials and some are under process like liposomes, SLNs, NLC, polymeric NP, micelle, dendrimers, etc. But the least numbers are actual in clinical trials for the lung cancer treatment and this could be the starting point to carry out novel delivery using nanotechnology. For the development of the novel delivery of these drugs we need to study the biology and microenvironment of the tumor and then interaction between prepared NP and tumor. Preparation of the NP simple, low cost, reproducible with high safety, selectivity and efficacious towards tumor. This study provides you with information about recent studies and future aspects of EGFR-tyrosine kinase receptor inhibitory drugs also its NPs and newer combinations that provide better therapy for NSCLC.

    Abbreviations

    DFS:

    disease-free survival

    EGFR:

    epidermal growth factor receptor

    NLC:

    nanostructured lipid carriers

    NPs:

    nanoparticles

    NSCLC:

    non-small cell lung cancer

    PEG:

    polyethylene glycol

    PLGA:

    polylactic-co-glycolic acid

    SCLC:

    small cell lung cancer

    SLN:

    solid lipid nanoparticle

    TKIs:

    tyrosine kinase inhibitors

    Declarations

    Acknowledgments

    Authors are thankful to the Management of Dr Vishwanath Karad MIT-World Peace University, Kothrud, Pune 411038.

    Author contributions

    PSW: Writing—original draft, Data curation. ARC: Conceptualization, Writing—review & editing. KGR: Data curation, Validation, Visualization. BGP: Writing—review & editing. SCJ: Supervision, Validation.

    Conflicts of interest

    The authors declare no conflicts of interest.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Availability of data and materials

    Not applicable.

    Funding

    Not applicable.

    Copyright

    © The Author(s) 2024.

    References

    Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394424. [DOI] [PubMed]
    Ferlay J, Colombet M, Soerjomataram I, Mathers C, Parkin DM, Piñeros M, et al. Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer. 2019;144:194153. [DOI] [PubMed]
    Wang J, Zhou T, Liu Y, Chen S, Yu Z. Application of Nanoparticles in the Treatment of Lung Cancer With Emphasis on Receptors. Front Pharmacol. 2022;12:781425. [DOI] [PubMed] [PMC]
    Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72:733. [DOI] [PubMed]
    Khanmohammadi A, Aghaie A, Vahedi E, Qazvini A, Ghanei M, Afkhami A, et al. Electrochemical biosensors for the detection of lung cancer biomarkers: A review. Talanta. 2020;206:120251. [DOI] [PubMed]
    Bethune G, Bethune D, Ridgway N, Xu Z. Epidermal growth factor receptor (EGFR) in lung cancer: an overview and update. J Thorac Dis. 2010;2:4851. [PubMed] [PMC]
    Jin X, Zhou J, Zhang Z, Lv H. The combined administration of parthenolide and ginsenoside CK in long circulation liposomes with targeted tLyp-1 ligand induce mitochondria-mediated lung cancer apoptosis. Artif Cells Nanomed Biotechnol. 2018;46:S93142. [DOI] [PubMed]
    Zappa C, Mousa SA. Non-small cell lung cancer: current treatment and future advances. Transl Lung Cancer Res. 2016;5:288300. [DOI] [PubMed] [PMC]
    Pao W, Girard N. New driver mutations in non-small-cell lung cancer. Lancet Oncol. 2011;12:17580. [DOI] [PubMed]
    Georgoulias V, Papadakis E, Alexopoulos A, Tsiafaki X, Rapti A, Veslemes M, et al.; Greek Oncology Cooperative Group (GOCG) for Lung Cancer. Platinum-based and non-platinum-based chemotherapy in advanced non-small-cell lung cancer: a randomised multicentre trial. Lancet. 2001;357:147884. [DOI] [PubMed]
    Yarden Y. The EGFR family and its ligands in human cancer. signalling mechanisms and therapeutic opportunities. Eur J Cancer. 2001;37:S38. [DOI] [PubMed]
    Benelmekki M. An introduction to nanoparticles and nanotechnology. In: Designing Hybrid Nanoparticles. Morgan & Claypool Publishers; 2015. pp. 1-1–14. [DOI]
    Almeida JPM, Lin AY, Langsner RJ, Eckels P, Foster AE, Drezek RA. In vivo immune cell distribution of gold nanoparticles in naïve and tumor bearing mice. Small. 2014;10:8129. [DOI] [PubMed] [PMC]
    Rosell R, Morán T, Carcereny E, Quiroga V, Molina MA, Costa C, et al. Non-small-cell lung cancer harbouring mutations in the EGFR kinase domain. Clin Transl Oncol. 2010;12:7580. [DOI] [PubMed]
    Bordi P, Re MD, Danesi R, Tiseo M. Circulating DNA in diagnosis and monitoring EGFR gene mutations in advanced non-small cell lung cancer. Transl Lung Cancer Res. 2015;4:58497. [DOI] [PubMed] [PMC]
    Yuan M, Huang L, Chen J, Wu J, Xu Q. The emerging treatment landscape of targeted therapy in non-small-cell lung cancer. Signal Transduct Target Ther. 2019;4:61. [DOI] [PubMed] [PMC]
    Lin C, Hu F, Chu H, Ren P, Ma S, Wang J, et al. The role of EGFR-TKIs as adjuvant therapy in EGFR mutation-positive early-stage NSCLC: A meta-analysis. Thorac Cancer. 2021;12:108495. [DOI] [PubMed] [PMC]
    Wu YL, Tsuboi M, He J, John T, Grohe C, Majem M, et al.; ADAURA Investigators. Osimertinib in Resected EGFR-Mutated Non-Small-Cell Lung Cancer. N Engl J Med. 2020;383:171123. [DOI] [PubMed]
    Zhong W, Wang Q, Mao W, Xu S, Wu L, Shen Y, et al.; ADJUVANT investigators. Gefitinib versus vinorelbine plus cisplatin as adjuvant treatment for stage II-IIIA (N1-N2) EGFR-mutant NSCLC (ADJUVANT/CTONG1104): a randomised, open-label, phase 3 study. Lancet Oncol. 2018;19:13948. [DOI] [PubMed]
    Yue D, Xu S, Wang Q, Li X, Shen Y, Zhao H, et al. Erlotinib versus vinorelbine plus cisplatin as adjuvant therapy in Chinese patients with stage IIIA EGFR mutation-positive non-small-cell lung cancer (EVAN): a randomised, open-label, phase 2 trial. Lancet Respir Med. 2018;6:86373. [DOI] [PubMed]
    Li N, Ou W, Ye X, Sun H, Zhang L, Fang Q, et al. Pemetrexed-carboplatin adjuvant chemotherapy with or without gefitinib in resected stage IIIA-N2 non-small cell lung cancer harbouring EGFR mutations: a randomized, phase II study. Ann Surg Oncol. 2014;21:20916. [DOI] [PubMed]
    Kelly K, Altorki NK, Eberhardt WE, O’Brien ME, Spigel DR, Crinò L, et al. Adjuvant Erlotinib Versus Placebo in Patients With Stage IB-IIIA Non-Small-Cell Lung Cancer (RADIANT): A Randomized, Double-Blind, Phase III Trial. J Clin Oncol. 2015;33:400714. [DOI] [PubMed]
    Sun L, Guo Y, Song J, Wang Y, Zhang S, Huang L, et al. Neoadjuvant EGFR-TKI Therapy for EGFR-Mutant NSCLC: A Systematic Review and Pooled Analysis of Five Prospective Clinical Trials. Front Oncol. 2021;10:586596. [DOI] [PubMed] [PMC]
    Sullivan I, Planchard D. Next-Generation EGFR Tyrosine Kinase Inhibitors for Treating EGFR-Mutant Lung Cancer beyond First Line. Front Med (Lausanne). 2017;3:76. [DOI] [PubMed] [PMC]
    Solassol I, Pinguet F, Quantin X. FDA- and EMA-Approved Tyrosine Kinase Inhibitors in Advanced EGFR-Mutated Non-Small Cell Lung Cancer: Safety, Tolerability, Plasma Concentration Monitoring, and Management. Biomolecules. 2019;9:668. [DOI] [PubMed] [PMC]
    Bodnar RJ. Anti-Angiogenic Drugs: Involvement in Cutaneous Side Effects and Wound-Healing Complication. Adv Wound Care (New Rochelle). 2014;3:63546. [DOI] [PubMed] [PMC]
    Özdemir A, Ciftci H, Sever B, Tateishi H, Otsuka M, Fujita M, et al. A New Series of Indeno[1,2- c]pyrazoles as EGFR TK Inhibitors for NSCLC Therapy. Molecules. 2022;27:485. [DOI] [PubMed] [PMC]
    TUSC2-nanoparticles and Erlotinib in Stage IV Lung Cancer [Internet]. National Library of Medicine; c2024 [cited 2022 Mar 2]. Available from: https://clinicaltrials.gov/study/NCT01455389
    FDA Grants Fast Track Designation to Quaratusugene Ozeplasmid Immunogene Therapy Plus Pembrolizumab for NSCLC [Internet]. Cranbury: MJH Life Sciences; c2024 [cited 2022 Jan 5]. Available from: https://www.cancernetwork.com/view/fda-grants-fast-track-designation-to-quaratusugene-ozeplasmid-immunogene-therapy-plus-pembrolizumab-for-nsclc
    Asahina H, Tanaka K, Morita S, Maemondo M, Seike M, Okamoto I, et al. A Phase II Study of Osimertinib Combined With Platinum Plus Pemetrexed in Patients With EGFR-Mutated Advanced Non-Small-cell Lung Cancer: The OPAL Study (NEJ032C/LOGIK1801). Clin Lung Cancer. 2021;22:14751. [DOI] [PubMed]
    Qin H, Wang F, Zeng Z, Jia S, Liu Y, Gao H. Effect of Osimertinib in Combination With Chemotherapy and Bevacizumab for Untreated Epidermal Growth Factor Receptor-Mutated Advanced Non-Small-Cell Lung Cancer: Case Report. Front Pharmacol. 2021;12:679667. [DOI] [PubMed] [PMC]
    Rotow JK, Costa DB, Paweletz CP, Awad MM, Marcoux P, Rangachari D, et al. Concurrent osimertinib plus gefitinib for first-line treatment of EGFR-mutated non-small cell lung cancer (NSCLC). J Clin Oncol. 2020;38:9507. [DOI]
    Solomon B, Mersiades A, Brown C, Dawson S, Wong S, Tan L, et al. P76.64 Alternating Osimertinib and Gefitinib as Second-Line Treatment for EGFR-Mutated NSCLC Harbouring a T790M Resistance Mutation (OSCILLATE). J Thorac Oncol. 2021;16:S6156. [DOI]
    Yang Y, Liu Q, Cao L, Sun W, Gu X, Liu B, et al. Osimertinib versus afatinib in patients with T790M-positive, non-small-cell lung cancer and multiple central nervous system metastases after failure of initial EGFR-TKI treatment. BMC Pulm Med. 2021;21:172. [DOI] [PubMed] [PMC]
    García-Pinel B, Porras-Alcalá C, Ortega-Rodríguez A, Sarabia F, Prados J, Melguizo C, et al. Lipid-Based Nanoparticles: Application and Recent Advances in Cancer Treatment. Nanomaterials (Basel). 2019;9:638. [DOI] [PubMed] [PMC]
    Markman JL, Rekechenetskiy A, Holler E, Ljubimova JY. Nanomedicine therapeutic approaches to overcome cancer drug resistance. Adv Drug Deliv Rev. 2013;65:186679. [DOI] [PubMed] [PMC]
    Zhang A, Meng K, Liu Y, Pan Y, Qu W, Chen D, et al. Absorption, distribution, metabolism, and excretion of nanocarriers in vivo and their influences. Adv Colloid Interface Sci. 2020;284:102261. [DOI] [PubMed]
    Wang B, He X, Zhang Z, Zhao Y, Feng W. Metabolism of nanomaterials in vivo: blood circulation and organ clearance. Acc Chem Res. 2013;46:7619. [DOI] [PubMed]
    Ravinayagam V, Jermy BR. Nanomaterials and their negative effects on human health. In: Khan F, editor. Applications of Nanomaterials in Human Health. Singapore: Springer; 2020. pp. 249–73. [DOI] [PMC]
    Patra JK, Das G, Fraceto LF, Campos EVR, Rodriguez-Torres MDP, Acosta-Torres LS, et al. Nano based drug delivery systems: recent developments and future prospects. J Nanobiotechnology. 2018;16:71. [DOI] [PubMed] [PMC]
    Sharma P, Mehta M, Dhanjal DS, Kaur S, Gupta G, Singh H, et al. Emerging trends in the novel drug delivery approaches for the treatment of lung cancer. Chem Biol Interact. 2019;309:108720. [DOI] [PubMed]
    Blanco E, Shen H, Ferrari M. Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat Biotechnol. 2015;33:94151. [DOI] [PubMed] [PMC]
    Liu Z, Tabakman S, Welsher K, Dai H. Carbon Nanotubes in Biology and Medicine: In vitro and in vivo Detection, Imaging and Drug Delivery. Nano Res. 2009;2:85120. [DOI] [PubMed] [PMC]
    Razzacki SZ, Thwar PK, Yang M, Ugaz VM, Burns MA. Integrated microsystems for controlled drug delivery. Adv Drug Deliv Rev. 2004;56:18598. [DOI] [PubMed]
    Orive G, Gascón AR, Hernández RM, Domínguez-Gil A, Pedraz JL. Techniques: new approaches to the delivery of biopharmaceuticals. Trends Pharmacol Sci. 2004;25:3827. [DOI] [PubMed]
    Kabanov AV, Lemieux P, Vinogradov S, Alakhov V. Pluronic block copolymers: novel functional molecules for gene therapy. Adv Drug Deliv Rev. 2002;54:22333. [DOI] [PubMed]
    Mirza AZ, Siddiqui FA. Nanomedicine and drug delivery: a mini review. Int Nano Lett. 2014;4:94. [DOI]
    Wang N, Feng Y. Elaborating the role of natural products-induced autophagy in cancer treatment: achievements and artifacts in the state of the art. Biomed Res Int. 2015;2015:934207. [DOI] [PubMed] [PMC]
    Carrasco-Esteban E, Domínguez-Rullán JA, Barrionuevo-Castillo P, Pelari-Mici L, Leaman O, Sastre-Gallego S, et al. Current role of nanoparticles in the treatment of lung cancer. J Clin Transl Res. 2021;7:14055. [PubMed] [PMC]
    Jong WHD, Borm PJA. Drug delivery and nanoparticles: applications and hazards. Int J Nanomedicine. 2008;3:13349. [DOI] [PubMed] [PMC]
    Lenaerts V, Nagelkerke JF, Berkel TJV, Couvreur P, Grislain L, Roland M, et al. In vivo uptake of polyisobutyl cyanoacrylate nanoparticles by rat liver Kupffer, endothelial, and parenchymal cells. J Pharm Sci. 1984;73:9802. [DOI] [PubMed]
    Bajpai S, Tiwary SK, Sonker M, Joshi A, Gupta V, Kumar Y, et al. Recent Advances in Nanoparticle-Based Cancer Treatment: A Review. ACS Appl Nano Mater. 2021;4:644170. [DOI]
    Zensi A, Begley D, Pontikis C, Legros C, Mihoreanu L, Wagner S, et al. Albumin nanoparticles targeted with Apo E enter the CNS by transcytosis and are delivered to neurones. J Control Release. 2009;137:7886. [DOI] [PubMed]
    Bazile D, Prud’homme C, Bassoullet MT, Marlard M, Spenlehauer G, Veillard M. Stealth Me.PEG-PLA nanoparticles avoid uptake by the mononuclear phagocytes system. J Pharm Sci. 1995;84:4938. [DOI] [PubMed]
    Amina SJ, Guo B. A Review on the Synthesis and Functionalization of Gold Nanoparticles as a Drug Delivery Vehicle. Int J Nanomedicine. 2020;15:982357. [DOI] [PubMed] [PMC]
    Panyam J, Zhou W, Prabha S, Sahoo SK, Labhasetwar V. Rapid endo-lysosomal escape of poly(DL-lactide-co-glycolide) nanoparticles: implications for drug and gene delivery. FASEB J. 2002;16:121726. [DOI] [PubMed]
    Konan YN, Chevallier J, Gurny R, Allémann E. Encapsulation of p-THPP into nanoparticles: cellular uptake, subcellular localization and effect of serum on photodynamic activity. Photochem Photobiol. 2003;77:63844. [DOI] [PubMed]
    Weissenböck A, Wirth M, Gabor F. WGA-grafted PLGA-nanospheres: preparation and association with Caco-2 single cells. J Control Release. 2004;99:38392. [DOI] [PubMed]
    Prinzen L, Miserus RJHM, Dirksen A, Hackeng TM, Deckers N, Bitsch NJ, et al. Optical and magnetic resonance imaging of cell death and platelet activation using annexin a5-functionalized quantum dots. Nano Lett. 2007;7:93100. [DOI] [PubMed]
    Nobs L, Buchegger F, Gurny R, Allémann E. Poly(lactic acid) nanoparticles labeled with biologically active Neutravidin for active targeting. Eur J Pharm Biopharm. 2004;58:48390. [DOI] [PubMed]
    Milenic DE, Brady ED, Brechbiel MW. Antibody-targeted radiation cancer therapy. Nat Rev Drug Discov. 2004;3:48899. [DOI] [PubMed]
    Zwicke GL, Mansoori GA, Jeffery CJ. Utilizing the folate receptor for active targeting of cancer nanotherapeutics. Nano Rev. 2012;3. [DOI] [PubMed] [PMC]
    Lo YL, Lin HC, Hong ST, Chang CH, Wang CS, Lin AMY. Lipid polymeric nanoparticles modified with tight junction-modulating peptides promote afatinib delivery across a blood–brain barrier model. Cancer Nanotechnol. 2021;12:13. [DOI]
    Skupin-Mrugalska P, Minko T. Development of Liposomal Vesicles for Osimertinib Delivery to EGFR Mutation-Positive Lung Cancer Cells. Pharmaceutics. 2020;12:939. [DOI] [PubMed] [PMC]
    Hu Y, Zhang J, Hu H, Xu S, Xu L, Chen E. Gefitinib encapsulation based on nano-liposomes for enhancing the curative effect of lung cancer. Cell Cycle. 2020;19:358194. [DOI] [PubMed] [PMC]
    Satari N, Taymouri S, Varshosaz J, Rostami M, Mirian M. Preparation and evaluation of inhalable dry powder containing glucosamine-conjugated gefitinib SLNs for lung cancer therapy. Drug Dev Ind Pharm. 2020;46:126577. [DOI] [PubMed]
    Zhang T, Wang R, Li M, Bao J, Chen Y, Ge Y, et al. Comparative study of intratracheal and oral gefitinib for the treatment of primary lung cancer. Eur J Pharm Sci. 2020;149:105352. [DOI] [PubMed]
    Chan M, Huang W, Wang J, Liu R, Hsiao M. Next-Generation Cancer-Specific Hybrid Theranostic Nanomaterials: MAGE-A3 NIR Persistent Luminescence Nanoparticles Conjugated to Afatinib for In Situ Suppression of Lung Adenocarcinoma Growth and Metastasis. Adv Sci (Weinh). 2020;7:1903741. [DOI] [PubMed] [PMC]
    Ganthala PD, Alavala S, Chella N, Andugulapati SB, Bathini NB, Sistla R. Co-encapsulated nanoparticles of Erlotinib and Quercetin for targeting lung cancer through nuclear EGFR and PI3K/AKT inhibition. Colloids Surf B Biointerfaces. 2022;211:112305. [DOI] [PubMed]
    Zhou X, He X, Shi K, Yuan L, Yang Y, Liu Q, et al. Injectable Thermosensitive Hydrogel Containing Erlotinib-Loaded Hollow Mesoporous Silica Nanoparticles as a Localized Drug Delivery System for NSCLC Therapy. Adv Sci (Weinh). 2020;7:2001442. [DOI] [PubMed] [PMC]
    Li K, Zhan W, Jia M, Zhao Y, Liu Y, Jha RK, et al. Dual Loading of Nanoparticles with Doxorubicin and Icotinib for the Synergistic Suppression of Non-Small Cell Lung Cancer. Int J Med Sci. 2020;17:390402. [DOI] [PubMed] [PMC]
    Pang J, Xing H, Sun Y, Feng S, Wang S. Non-small cell lung cancer combination therapy: Hyaluronic acid modified, epidermal growth factor receptor targeted, pH sensitive lipid-polymer hybrid nanoparticles for the delivery of erlotinib plus bevacizumab. Biomed Pharmacother. 2020;125:109861. [DOI] [PubMed]
    Wang D, Zhou J, Fang W, Huang C, Chen Z, Fan M, et al. A multifunctional nanotheranostic agent potentiates erlotinib to EGFR wild-type non-small cell lung cancer. Bioact Mater. 2021;13:31223. [DOI] [PubMed] [PMC]
    Upadhyay P, Ghosh A, Basu A, Pranati PA, Gupta P, Das S, et al. Delivery of gefitinib in synergism with thymoquinone via transferrin-conjugated nanoparticle sensitizes gefitinib-resistant non-small cell lung carcinoma to control metastasis and stemness. Biomater Sci. 2021;9:8285312. [DOI] [PubMed]
    Chen W, Yu D, Sun S, Li F. Nanoparticles for co-delivery of osimertinib and selumetinib to overcome osimertinib-acquired resistance in non-small cell lung cancer. Acta Biomater. 2021;129:25868. [DOI] [PubMed] [PMC]
    Lee Y, Lee D, Park E, Jang S, Cheon SY, Han S, et al. Rhamnolipid-coated W/O/W double emulsion nanoparticles for efficient delivery of doxorubicin/erlotinib and combination chemotherapy. J Nanobiotechnology. 2021;19:411. [DOI] [PubMed] [PMC]
    Bozzuto G, Molinari A. Liposomes as nanomedical devices. Int J Nanomedicine. 2015;10:97599. [DOI] [PubMed] [PMC]
    Sercombe L, Veerati T, Moheimani F, Wu SY, Sood AK, Hua S. Advances and Challenges of Liposome Assisted Drug Delivery. Front Pharmacol. 2015;6:286. [DOI] [PubMed] [PMC]
    Yingchoncharoen P, Kalinowski DS, Richardson DR. Lipid-Based Drug Delivery Systems in Cancer Therapy: What Is Available and What Is Yet to Come. Pharmacol Rev. 2016;68:70187. [DOI] [PubMed] [PMC]
    Lu X, Liu S, Han M, Yang X, Sun K, Wang H, et al. Afatinib-loaded immunoliposomes functionalized with cetuximab: A novel strategy targeting the epidermal growth factor receptor for treatment of non-small-cell lung cancer. Int J Pharm. 2019;560:12635. [DOI] [PubMed]
    Lv X, Yin J, Yang X, Liu S, Sun K. [Formulation and Efficacy of Liposome-encapsulated Afatinib for Therapy of Non-small Cell Lung Cancer]. Zhongguo Fei Ai Za Zhi. 2018;21:6639. Chinese. [DOI] [PubMed] [PMC]
    Almurshedi AS, Radwa M, Omar S, Alaiya AA, Badran MM, Elsaghire H, et al. A novel pH-sensitive liposome to trigger delivery of afatinib to cancer cells: Impact on lung cancer therapy. J Mol Liq. 2018;259:15466. [DOI]
    Paliwal R, Paliwal SR, Kenwat R, Kurmi BD, Sahu MK. Solid lipid nanoparticles: a review on recent perspectives and patents. Expert Opin Ther Pat. 2020;30:17994. [DOI] [PubMed]
    Andreozzi E, Seo JW, Ferrara K, Louie A. Novel method to label solid lipid nanoparticles with 64cu for positron emission tomography imaging. Bioconjug Chem. 2011;22:80818. [DOI] [PubMed] [PMC]
    Pan X, Zhang X, Sun H, Zhang J, Yan M, Zhang H. Autophagy Inhibition Promotes 5-Fluorouraci-Induced Apoptosis by Stimulating ROS Formation in Human Non-Small Cell Lung Cancer A549 Cells. PLoS ONE. 8:e56679. [DOI] [PubMed] [PMC]
    Bakhtiary Z, Barar J, Aghanejad A, Saei AA, Nemati E, Dolatabadi JEN, et al. Microparticles containing erlotinib-loaded solid lipid nanoparticles for treatment of non-small cell lung cancer. Drug Dev Ind Pharm. 2017;43:124453. [DOI] [PubMed]
    Gaur PK, Mishra S, Purohit S. Solid lipid nanoparticles of guggul lipid as drug carrier for transdermal drug delivery. Biomed Res Int. 2013;2013:750690. [DOI] [PubMed] [PMC]
    Zielińska W, Nawrocka A, Rydzkowski M, Kokocha A, Matulewicz K, Hałas-Wiśniewska M, et al. Possibilities in the application of solid lipid nanoparticles in combination with 5-fluorouracil to overcome the drugresistance of non-small cell lung cancer cell line A549. Med Res J. 2020;5:18. [DOI]
    Duan Y, Dhar A, Patel C, Khimani M, Neogi S, Sharma P, et al. A brief review on solid lipid nanoparticles: part and parcel of contemporary drug delivery systems. RSC Adv. 2020;10:2677791. [DOI] [PubMed] [PMC]
    Makled S, Nafee N, Boraie N. Nebulized solid lipid nanoparticles for the potential treatment of pulmonary hypertension via targeted delivery of phosphodiesterase-5-inhibitor. Int J Pharm. 2017;517:31221. [DOI] [PubMed]
    Yang Y, Huang Z, Li J, Mo Z, Huang Y, Ma C, et al. PLGA Porous Microspheres Dry Powders for Codelivery of Afatinib-Loaded Solid Lipid Nanoparticles and Paclitaxel: Novel Therapy for EGFR Tyrosine Kinase Inhibitors Resistant Nonsmall Cell Lung Cancer. Adv Healthc Mater. 2019;8:e1900965. [DOI] [PubMed]
    Haider M, Abdin SM, Kamal L, Orive G. Nanostructured Lipid Carriers for Delivery of Chemotherapeutics: A Review. Pharmaceutics. 2020;12:288. [DOI] [PubMed] [PMC]
    Khosa A, Reddi S, Saha RN. Nanostructured lipid carriers for site-specific drug delivery. Biomed Pharmacother. 2018;103:598613. [DOI] [PubMed]
    Ghasemiyeh P, Mohammadi-Samani S. Solid lipid nanoparticles and nanostructured lipid carriers as novel drug delivery systems: applications, advantages and disadvantages. Res Pharm Sci. 2018;13:288303. [DOI] [PubMed] [PMC]
    Hoffman AS. The origins and evolution of “controlled” drug delivery systems. J Control Release. 2008;132:15363. [DOI] [PubMed]
    Chan JM, Valencia PM, Zhang L, Langer R, Farokhzad OC. Polymeric nanoparticles for drug delivery. Methods Mol Biol. 2010;624:16375. [DOI] [PubMed]
    Singh S, Nalwa HS. Nanotechnology and health safety--toxicity and risk assessments of nanostructured materials on human health. J Nanosci Nanotechnol. 2007;7:304870. [DOI] [PubMed]
    Vaidya B, Parvathaneni V, Kulkarni NS, Shukla SK, Damon JK, Sarode A, et al. Cyclodextrin modified erlotinib loaded PLGA nanoparticles for improved therapeutic efficacy against non-small cell lung cancer. Int J Biol Macromol. 2019;122:33847. [DOI] [PubMed]
    Nikolova C, Gutierrez T. Biosurfactants and Their Applications in the Oil and Gas Industry: Current State of Knowledge and Future Perspectives. Front Bioeng Biotechnol. 2021;9:626639. [DOI] [PubMed] [PMC]
    Xu W, Ling P, Zhang T. Polymeric micelles, a promising drug delivery system to enhance bioavailability of poorly water-soluble drugs. J Drug Deliv. 2013;2013:340315. [DOI] [PubMed] [PMC]
    Miyata K, Christie RJ, Kataoka K. Polymeric micelles for nano-scale drug delivery. React Funct Polym. 2011;71:22734. [DOI]
    Zhou X, Shi K, Hao Y, Yang C, Zha R, Yi C, et al. Advances in nanotechnology-based delivery systems for EGFR tyrosine kinases inhibitors in cancer therapy. Asian J Pharm Sci. 2020;15:2641. [DOI] [PubMed] [PMC]
    Sandoval-Yañez C, Rodriguez CC. Dendrimers: Amazing Platforms for Bioactive Molecule Delivery Systems. Materials (Basel). 2020;13:570. [DOI] [PubMed] [PMC]
    Sharma AK, Gothwal A, Kesharwani P, Alsaab H, Iyer AK, Gupta U. Dendrimer nanoarchitectures for cancer diagnosis and anticancer drug delivery. Drug Discov Today. 2017;22:31426. [DOI] [PubMed]
    Chis AA, Dobrea C, Morgovan C, Arseniu AM, Rus LL, Butuca A, et al. Applications and Limitations of Dendrimers in Biomedicine. Molecules. 2020;25:3982. [DOI] [PubMed] [PMC]
    Mohtar N, Parumasivam T, Gazzali AM, Tan CS, Tan ML, Othman R, et al. Advanced Nanoparticle-Based Drug Delivery Systems and Their Cellular Evaluation for Non-Small Cell Lung Cancer Treatment. Cancers (Basel). 2021;13:3539. [DOI] [PubMed] [PMC]
    Issa B, Obaidat IM, Albiss BA, Haik Y. Magnetic nanoparticles: surface effects and properties related to biomedicine applications. Int J Mol Sci. 2013;14:21266305. [DOI] [PubMed] [PMC]
    Oerlemans C, Bult W, Bos M, Storm G, Nijsen JFW, Hennink WE. Polymeric micelles in anticancer therapy: targeting, imaging and triggered release. Pharm Res. 2010;27:256989. [DOI] [PubMed] [PMC]
    Yu AYH, Fu RH, Hsu SH, Chiu CF, Fang WH, Yeh CA, et al. Epidermal growth factor receptors siRNA-conjugated collagen modified gold nanoparticles for targeted imaging and therapy of lung cancer. Mater Today Adv. 2021;12:100191. [DOI]
    Saifuddin N, Raziah AZ, Junizah AZ. Carbon Nanotubes: A Review on Structure and Their Interaction with Proteins. J Chem. 2013;2013:676815. [DOI]
    Casals E, Puntes VF. Inorganic nanoparticle biomolecular corona: formation, evolution and biological impact. Nanomedicine (Lond). 2012;7:191730. [DOI] [PubMed]
    Barbarino M, Giordano A. Assessment of the Carcinogenicity of Carbon Nanotubes in the Respiratory System. Cancers (Basel). 2021;13:1318. [DOI] [PubMed] [PMC]
    Li X, Wang L, Fan Y, Feng Q, Cui FZ. Biocompatibility and toxicity of nanoparticles and nanotubes. J Nanomater. 2012;2012:548389. [DOI]