Exploiting pH-Sensitive Polymer Micelles Nanoparticles for Paclitaxel Delivery and Tumor Suppression: Advanced Targeted Cancer Therapy

Document Type : Research Paper

Authors

1 Department of Polymer Chemistry and Chemical Technology, Samarkand state university, Samarkand, Uzbekistan

2 College of Pharmacy, Ahl Al Bayt University, Kerbala, Iraq

3 AlSafwa University College, Iraq

4 Department of Pharmaceutics, College of Pharmacy, University of Al-Ameed, Karbala, Iraq

5 Department of sciences, Al-Manara College for Medical Sciences, Maysan, Iraq

6 Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq

7 Mazaya University College, Iraq

8 Al-Zahrawi University College, Karbala, Iraq

9 Department Clinic Pharmacology, Bukhara State Medical Institute, Uzbekistan

10 Department of Fruits and Vegetables, Urganch State University, Uzbekistan

11 Department of Neurology and Psychiatry, Fergana Medical Institute of Public Health, Fergana, Uzbekistan

12 Samarkand State Veterinary Medicine, Nukus branch, Livestock and biotechnologies, Uzbekistan

13 International School of Finance Technology and Science, Uzbekistan

10.22052/JNS.2024.03.022

Abstract

Targeted cancer therapy aims to enhance the efficacy and safety of treatments by directing therapeutic agents specifically to tumor sites. This review discusses the potential of pH-sensitive polymer micelles as carriers for paclitaxel, a chemotherapeutic agent, in advanced cancer treatment. By exploiting the unique tumor microenvironment, these nanoparticles can effectively release paclitaxel in a controlled manner, enhancing drug accumulation in cancer cells while minimizing systemic toxicity. The combination of paclitaxel with pH-sensitive polymer micelles presents a promising strategy for tumor suppression, potentially overcoming the limitations of conventional chemotherapy. The review highlights recent advancements, mechanisms of action, and clinical implications of using pH-sensitive polymer micelles for paclitaxel delivery.

Keywords


INTRODUCTION
Cancer remains one of the foremost challenges in modern medicine, with its complex and heterogeneous nature complicating treatment strategies. Traditional chemotherapy, which targets rapidly dividing cells, often lacks the specificity needed to distinguish between cancerous and healthy cells [1]. This non-selective approach can lead to significant adverse effects, including damage to vital organs, hair loss, and severe fatigue, which can compromise patient quality of life [2]. In recent years, targeted cancer therapy has emerged as a promising alternative, aiming to deliver therapeutic agents directly to cancer cells while sparing healthy tissues [3]. This approach leverages the unique biological markers and microenvironment of tumors to achieve a higher therapeutic index. Methods such as receptor-mediated targeting, using monoclonal antibodies, and small-molecule inhibitors have demonstrated significant potential in improving the specificity and efficacy of cancer treatments [4]. Nanoparticles have revolutionized the field of drug delivery due to their ability to enhance the pharmacokinetic and pharmacodynamic profiles of therapeutic agents. These nanoscale carriers, typically ranging from 10 to 100 nanometers in size, can be engineered to exhibit unique properties such as prolonged circulation time, enhanced permeability and retention (EPR) effect, and the ability to bypass biological barriers [5]. Nanoparticles can be designed to deliver a wide range of therapeutic agents, including small molecules, proteins, and nucleic acids. They offer several advantages over conventional drug delivery systems, such as improved solubility of hydrophobic drugs, controlled and sustained release, and targeted delivery to specific tissues or cells. These properties not only enhance the efficacy of the therapeutic agents but also minimize off-target effects, thereby reducing systemic toxicity [6]. pH-sensitive polymer micelles have garnered significant interest in the realm of targeted drug delivery due to their ability to respond to the acidic tumor microenvironment. These micelles are composed of amphiphilic block copolymers that self-assemble into core-shell structures in aqueous solutions [7]. The hydrophobic core serves as a reservoir for the encapsulated drug, while the hydrophilic shell provides stability and prolongs circulation time in the bloodstream [8]. The design of pH-sensitive polymer micelles takes advantage of the acidic pH found in tumor tissues (pH 6.5-6.8) compared to normal physiological pH (7.4). This pH differential can trigger the micelles to undergo structural changes or disassemble, releasing their drug payload specifically within the tumor microenvironment [9]. This targeted release mechanism ensures that the therapeutic agent is concentrated at the site of action, thereby maximizing its efficacy and minimizing systemic side effects [10]. The efficacy of pH-sensitive polymer micelles in cancer therapy is attributed to their ability to exploit the acidic conditions of the tumor microenvironment. Upon encountering the lower pH of tumor tissues, these micelles release their drug cargo in a controlled and sustained manner, enhancing drug accumulation within cancer cells. This approach not only improves the therapeutic index of the encapsulated drug but also reduces the likelihood of resistance development, as the localized high concentration of the drug can effectively eradicate tumor cells[11]. Paclitaxel, a potent anticancer agent, is widely used in the treatment of various cancers, including breast, ovarian, and lung cancers. However, its clinical application is limited by poor water solubility, low bioavailability, and significant systemic toxicity [12]. Incorporating paclitaxel into pH-sensitive polymer micelles addresses these challenges by enhancing its solubility, stability, and targeted delivery to tumor sites. The combination of paclitaxel with pH-sensitive polymer micelles offers several advantages, including improved therapeutic efficacy, reduced side effects, and the potential for overcoming multidrug resistance. This synergistic approach leverages the strengths of both components, providing a robust platform for advanced targeted cancer therapy [13].
   This review aims to explore the potential of pH-sensitive polymer micelles as carriers for paclitaxel delivery in cancer therapy. We will examine the mechanisms of action, recent advancements in the field, and the clinical implications of this innovative approach.
The novelty of this review lies in its comprehensive analysis of the synergistic effects of paclitaxel and pH-sensitive polymer micelles in targeted cancer therapy.

OVERVIEW OF PACLITAXEL 
Mechanism of Action
Paclitaxel, a member of the taxane family, operates by binding to the β-tubulin subunit of microtubules, thereby stabilizing them and preventing their depolymerisation [14]. This disruption of microtubule dynamics leads to mitotic arrest and the induction of apoptosis. The drug’s ability to inhibit cell division makes it effective against rapidly dividing cancer cells [15]. Research has shown that paclitaxel not only promotes the assembly of microtubules from tubulin dimers but also stabilizes them against depolymerization, causing the formation of abnormal bundles of microtubules throughout the cell cycle and multiple microtubule organizing centers during mitosis [16]. This interruption in the mitotic spindle assembly, chromosome segregation, and cell division results in cell death. New findings suggest that paclitaxel may also activate various apoptotic pathways, such as the caspase pathway, leading to programmed cell death in cancer cells [17]. Additionally, paclitaxel has been found to modulate the tumor microenvironment, including the inhibition of angiogenesis (the formation of new blood vessels), which is crucial for tumor growth and metastasis [18].

Current Challenges in Delivery
Despite its efficacy, paclitaxel faces several significant challenges in clinical use:
Paclitaxel is highly lipophilic, making it poorly soluble in water. This necessitates the use of solvents like Cremophor EL, which can cause severe hypersensitivity reactions, including anaphylaxis [19]. The high systemic toxicity of paclitaxel limits the doses that can be safely administered, leading to significant side effects such as neutropenia (a low count of neutrophils, a type of white blood cell), peripheral neuropathy (damage to the peripheral nerves causing pain and numbness), and cardiotoxicity [20].
Over time, cancer cells can develop resistance to paclitaxel through various mechanisms, such as the overexpression of P-glycoprotein, which pumps the drug out of the cells, or mutations in β-tubulin that prevent paclitaxel binding. New research is focusing on understanding these resistance mechanisms and developing strategies to overcome them, such as combination therapies and the use of inhibitors to block resistance pathways [21]. Clinical Applications of Paclitaxel is a cornerstone in the treatment of various cancers, including Ovarian Cancer, Breast Cancer, Non-Small Cell Lung Cancer, Kaposi’s Sarcoma, and Pancreatic Cancer [13].

pH-SENSITIVE POLYMER MICELLES
Types of Polymer Micelles Nanoparticles
pH-sensitive polymer micelles are a class of nanocarriers designed to enhance drug delivery to tumors by exploiting the acidic microenvironment characteristic of many cancerous tissues [22]. These nanoparticles can be broadly categorized based on their composition and structural attributes:
Block Copolymer Micelles: Composed of amphiphilic block copolymers, these micelles consist of hydrophobic cores and hydrophilic shells. The hydrophobic core can encapsulate hydrophobic drugs like paclitaxel, while the hydrophilic shell provides stability in aqueous environments [23]. Examples include poly(ethylene glycol)-block-poly(ε-caprolactone) (PEG-b-PCL) and poly(ethylene glycol)-block-poly(D,L-lactide) (PEG-b-PLA) [24].
Core-Shell Micelles: These micelles have a distinct core-shell structure, with the core designed to carry the drug and the shell providing steric stabilization. The core can be functionalized with pH-sensitive linkages that degrade in acidic environments, releasing the drug at the tumor site [11].
Polyion Complex (PIC) Micelles: Formed by the electrostatic interaction between oppositely charged polymers, PIC micelles can encapsulate both hydrophilic and hydrophobic drugs. They are particularly useful for delivering nucleic acids and proteins [25].

Design and Synthesis
The design and synthesis of pH-sensitive polymer micelles involve several critical steps to ensure their stability, biocompatibility, and responsiveness to pH changes [26], The choice of polymers is crucial for the micelle’s performance. Polymers with pH-sensitive functional groups, such as poly(2-(diisopropylamino)ethyl methacrylate) (PDPA) or poly(β-amino esters), are commonly used. These polymers undergo conformational changes or degrade at acidic pH, triggering drug release [27]. Micelles are typically formed by self-assembly of amphiphilic block copolymers in an aqueous solution. This process can be driven by solvent evaporation, dialysis, or nanoprecipitation methods. During self-assembly, the hydrophobic segments form the core, while the hydrophilic segments form the shell [28]. The encapsulation of drugs within the micelles can be achieved through physical entrapment or chemical conjugation. Physical entrapment involves dissolving the drug and polymer in a common solvent followed by self-assembly. Chemical conjugation involves attaching the drug to the polymer via pH-sensitive linkages, ensuring controlled release in the acidic tumor environment [29]. Comprehensive characterization of the micelles is essential to ensure their efficacy. Techniques such as dynamic light scattering (DLS) for size measurement, transmission electron microscopy (TEM) for morphological analysis, and high-performance liquid chromatography (HPLC) for drug loading efficiency are commonly used [30].

Mechanism of pH-Responsiveness
The pH-responsiveness of polymer micelles is attributed to the presence of pH-sensitive functional groups within the polymer structure. These groups undergo chemical or physical changes in response to acidic pH, leading to the release of the encapsulated drug [31], Polymers containing ionizable groups, such as carboxylic acids or amines, can alter their ionization state in response to pH changes. For instance, PDPA becomes protonated in acidic environments, leading to micelle destabilization and drug release [32]. The inclusion of pH-sensitive linkages, such as acetal, hydrazone, or orthoester bonds, in the polymer backbone allows for controlled degradation in acidic environments. These linkages remain stable at physiological pH but cleave in the acidic tumor microenvironment, releasing the drug [33]. Some polymers undergo conformational changes in response to pH variations. For example, poly(L-histidine) (PLH)-based micelles can transition from a coil to a globule state in acidic conditions, promoting the release of the drug [34].

Mechanism of Action with Paclitaxel
pH-sensitive polymer micelles are designed to enhance the delivery of paclitaxel to tumor sites. These micelles are stable at physiological pH but become destabilized in the acidic environment of tumor tissues, leading to the controlled release of paclitaxel directly at the tumor site [22]. The acidic pH of the tumor microenvironment (typically around pH 6.5 or lower) triggers the release of the encapsulated drug, improving the local concentration of paclitaxel and thereby enhancing its anticancer effects [35]. Recent studies have shown that pH-sensitive micelles can improve the pharmacokinetics of paclitaxel, leading to prolonged circulation time and increased accumulation in tumor tissues [31]. This is achieved through the enhanced permeability and retention (EPR) effect, where nanoparticles preferentially accumulate in tumor tissues due to the leaky vasculature and poor lymphatic drainage [36]. Furthermore, pH-sensitive micelles can be engineered to release their payload in response to specific triggers, such as enzymatic activity or changes in the redox environment, providing additional control over drug release and minimizing off-target effects [37].

DRUG LOADING AND RELEASE MECHANISMS
Strategies for Paclitaxel Loading
Effective drug loading is crucial for the success of pH-sensitive polymer micelles in delivering paclitaxel. Various strategies are employed to achieve high loading efficiency and stability [38].  Physical Encapsulation method involves dissolving both the drug and the polymer in a common organic solvent, followed by self-assembly into micelles through processes such as solvent evaporation or nanoprecipitation. Paclitaxel is physically entrapped within the hydrophobic core of the micelles, which enhances its solubility and stability [39]. Chemical Conjugation that in this approach, paclitaxel is covalently bonded to the polymer via pH-sensitive linkages such as hydrazone, acetal, or disulfide bonds. These linkages remain stable at physiological pH but cleave in the acidic tumor microenvironment, releasing the drug in a controlled manner. This strategy can significantly enhance the loading capacity and release control, minimizing premature drug release and systemic toxicity [40]. Core-Shell Micelle Formation whit utilizing block copolymers, are formed with distinct core-shell structures. The hydrophobic core encapsulates paclitaxel, while the hydrophilic shell stabilizes the micelles in aqueous environments. The core can be tailored to enhance drug loading by modifying the polymer composition and structure [41]. With Prodrug Approach, Paclitaxel can be modified into a prodrug that is more easily encapsulated by the micelles. Once the micelles reach the tumor site, the prodrug is activated by enzymatic or chemical triggers specific to the tumor environment, releasing the active drug [42].

pH-Dependent Release Dynamics
The release of paclitaxel from pH-sensitive polymer micelles is governed by the acidic conditions of the tumor microenvironment. Several mechanisms contribute to this controlled release [38], included Ionization of Polymer Chain which Polymers containing ionizable groups, such as carboxyl or amino groups, can undergo protonation or deprotonation in response to pH changes. At acidic pH, these groups become protonated, causing the micelle structure to destabilize and release the encapsulated drug. For instance, poly(β-amino ester) micelles can release paclitaxel in response to the acidic pH of the tumor site [40]. Cleavage of pH-Sensitive Bonds that pH-sensitive linkages within the polymer backbone, such as hydrazone, acetal, or orthoester bonds, are designed to break under acidic conditions. This cleavage triggers the release of paclitaxel from the micelle core. For example, hydrazone bonds are stable at neutral pH but hydrolyze in acidic environments, releasing the drug [43]. Some pH-sensitive polymers can swell in response to acidic pH, leading to an increase in the micelle size and porosity. This swelling facilitates the diffusion of paclitaxel out of the micelle core. Polymers like poly(L-histidine) can undergo such swelling, enhancing drug release in the acidic tumor microenvironment [44]. Enhanced Permeability and Retention (EPR) Effect: The EPR effect plays a significant role in the accumulation of pH-sensitive polymer micelles in tumor tissues. Due to the leaky vasculature and poor lymphatic drainage of tumors, micelles preferentially accumulate at the tumor site [45]. The acidic pH of the tumor further triggers the release of paclitaxel, enhancing its therapeutic efficacy. Studies have shown that micelles with pH-sensitive linkages exhibit enhanced drug release and antitumor activity compared to non-responsive micelles [46].

COMBINATION OF PACLITAXEL AND PH-SENSITIVE POLYMER MICELLES IN CANCER THERAPY 
Advantages of Paclitaxel-Loaded pH-Sensitive Micelles
The encapsulation of Paclitaxel within the hydrophobic core of pH-sensitive polymer micelles eliminates the need for toxic solubilizers such as Cremophor EL [47]. This significantly enhances patient safety by reducing adverse reactions commonly associated with these solubilizers. The pH-sensitive nature of these micelles ensures minimal drug release in normal tissues, thereby reducing systemic side effects such as neutropenia and neurotoxicity [39, 48]. The drug release mechanism is activated specifically in the acidic tumor microenvironment, thereby maximizing therapeutic efficacy and concentrating the drug’s action where it is needed most [49]. Paclitaxel-loaded micelles exhibit prolonged circulation time and preferential accumulation in tumors through the enhanced permeability and retention (EPR) effect [50]. This targeted delivery system significantly reduces off-target toxicity while maintaining high drug concentrations within tumor.This ensures that healthy cells are spared from the drug’s toxic effects, leading to a better overall safety profile [12]. In drug-resistant cancer cells, the acidic environment further enhances the release of Paclitaxel directly into the cytoplasm [51]. This targeted release bypasses drug efflux mechanisms mediated by P-glycoproteins, overcoming one of the major challenges in cancer therapy and improving the drug’s effectiveness against resistant cancer strains [52].

Clinical Studies 
Some findings underscore the importance of designing micelles with specific properties to optimize drug delivery and efficacy.  Yanhua Jiang et al [53], Conducted a study, pH-sensitive polyprodrug turned into used as nanocarrier, and PTX become encapsulated into the micelles with excessive drug-loading content (25.6%). The essential micelle concentration (CMC) become approximately 3.60 mg/L, indicating the machine should shape the micelles at low awareness. The particle length of PTX/DOX-PMs was 110.5 nm, and extended to about 140 nm after incubation for five days which confirmed that the PTX/DOX-PMs had excessive serum stability. With lower in pH price, the particle size first improved, and thenwas no longer detectable. Comparable exchange trend changed into observed for CMC values. The zetapotential improved sharply with decrease in pH. These effects validated the pHsensitivity of PTX/DOX-PMs. In vitro drug release experiments and study on release mechanism showed that the drug launch charge and accumulative launch for PTX and DOX were depending on the pH, displaying the pH-triggered drug release profiles. Cytotoxicity assay displayed that the block copolymer confirmed negligible cytotoxicity, even as the PTX/DOX-PMs possessed excessive cytotoxic impact against numerous tumor cellular traces compared with free drugs and manage.
Yang Han et al [54], studied amphiphilic conjugate based on mPEG and cholesterol-modified chitosan with hydrazone bonds within the molecules (mPEG-CS-Hz-CH) changed into effectively synthesized. the use of the polymer because the carrier, the paclitaxel (PTX)-loaded mPEG-CS-Hz-CH micelles had been organized by means of an ultrasonic probe method. The imply particle length and zeta ability of the optimized PTX-loaded micelles have been 146 ± 4 nm and +21.7 ± 0.7 mV, respectively. An in vitro drug release observe indicated that the PTX-loaded mPEG-CS-Hz-CH micelles had been solid underneath normal physiological conditions (pH 7.4), whereas speedy drug release become observed within the simulated tumor intracellular microenvironment (pH 5.0). An in vitro cytotoxicity have a look at demonstrated the non-toxicity of the polymer itself, and the PTX-loaded micelles exhibited superior cytotoxicity and huge selectivity on tumor cells. An in vivo antitumor efficacy examine similarly confirmed that the PTX-loaded micelles ought to enhance the therapeutic efficacy of PTX and decrease the aspect consequences. all these outcomes recommended that the mPEG-CS-Hz-CH micelles might be promising pH-sensitive nanocarriers for PTX delivery.
Peilan Qi et al [55], Presented a diblock copolymer constituting of a poly(ethylene glycol) (PEG) and a polycaprolactone (PCL) phase related through a pH-touchy hydrazone bond (Hyd), which become denoted as mPEG-Hyd-PCL, become synthesized. The copolymer became assembled to micelles with imply diameters about one hundred nm. The suggest diameters and size distribution of the hydrazone-containing micelles multiplied glaringly in mildly acidic environments while kept unchanged in the neutral. No tremendous change in size become observed on polymeric micelles without hydrazone (mPEG-PCL). PTX turned into loaded into micelles, and the anticancer drug released from mPEG-Hyd-PCL micelles become promoted by using the extended acidity. In vitro cytotoxicity examine showed that the PTX-loaded mPEG-Hyd-PCL micelles exhibited appreciably more suitable cytotoxicity towards HepG2 cells compared to the non-sensitive mPEG-PCL micelles. These consequences suggest that hydrazone-containing copolymer micelles with pH sensitivity and biodegradability display first rate ability as companies of anticancer drugs.
Jiajia Xiang et al [56], established a facile fabrication of a solid PTX-binding micelle crafted from poly (ethylene glycol)-block-dendritic polylysine, whose number one amines were reacted with phenethyl isothiocyanate (PEITC), a hydrophobic anticancer agent under clinical study. The amphiphilic conjugate (PEG-Gx-PEITC; Gx, the era of the polylysine dendron) fashioned properly-described micelles whose middle turned into composed of phenyl businesses and thiourea organizations binding PTX via π-π stacking and hydrogen bonding. as compared with the PTX-loaded poly(ethylene glycol)-block-poly(D,L-lactide) (PEGPDLLA/ PTX) micelles in scientific use, PTX-loaded PEG-Gx-PEITC 0.33-generation (PEG-G3-PEITC/PTX) micelles showed slowed blood clearance, enhanced tumor accumulation, and accordingly much advanced in vivo therapeutic efficacy in both subcutaneous and orthotopic human breast cancer xenografts. Consequently, PEG-G3-PEITC is a promising drug delivery machine for PTX inside the remedy of breast cancer.

CONCLUSION
The integration of pH-sensitive polymer micelles for paclitaxel delivery represents a significant advancement in targeted cancer therapy. By leveraging the acidic tumor microenvironment, these smart nanoparticles enable precise and controlled drug release, resulting in enhanced therapeutic efficacy and reduced systemic side effects. Recent advancements in micelle design, such as dual-targeting capabilities and co-delivery systems, have demonstrated their potential to overcome multidrug resistance and improve clinical outcomes in patients with aggressive or resistant tumors. The ability of pH-sensitive polymer micelles to enhance paclitaxel accumulation within cancer cells while sparing healthy tissues underscores their importance as a next-generation drug delivery platform in oncology.
Despite their potential, several challenges remain for the clinical translation of pH-sensitive polymer micelles. Scalable and cost-effective production methods are needed to ensure consistent quality and stability. Enhancements in targeting specificity, such as the incorporation of ligands for tumor-specific biomarkers, could further improve therapeutic precision. Research on combining pH-sensitive micelles with other treatment modalities, such as immunotherapies or gene therapies, could expand their application and effectiveness. Incorporating imaging agents for theranostic purposes could also support personalized treatment strategies. Finally, comprehensive safety and regulatory evaluations are necessary to confirm their suitability for clinical use. Future efforts should focus on optimizing these systems for clinical application, making pH-sensitive polymer micelles a key tool in advancing cancer therapy.

CONFLICT OF INTEREST
The authors declare that there is no conflict of interests regarding the publication of this manuscript.

1.    Kumar M. Targeting Cancer Cell Signaling Using Precision Oncology Towards a Holistic Approach to Cancer Therapeutics. Qeios Ltd; 2023.
2.    Messin C, Amrhein C. Evidence-Based Nursing in Basic Anticancer Treatment: Management of the Most Important Side Effects. Principles of Specialty Nursing: Springer International Publishing; 2019. p. 39-64.
3.    Reddy KTK, Reddy AS. Recent breakthroughs in drug delivery systems for targeted cancer therapy: an overview. Cellular, Molecular and Biomedical Reports. 2025;5(1):13-27.
4.    Sah N, Shaik AA, Acharya G, Dunna M, Silwal A, Sharma S, et al. Receptor-Based Strategies for Overcoming Resistance in Cancer Therapy. Receptors. 2024;3(4):425-443.
5.    Abdellatif AAH, Mohammed HA, Khan RA, Singh V, Bouazzaoui A, Yusuf M, et al. Nano-scale delivery: A comprehensive review of nano-structured devices, preparative techniques, site-specificity designs, biomedical applications, commercial products, and references to safety, cellular uptake, and organ toxicity. Nanotechnology Reviews. 2021;10(1):1493-1559.
6.    Mitchell MJ, Billingsley MM, Haley RM, Wechsler ME, Peppas NA, Langer R. Engineering precision nanoparticles for drug delivery. Nature reviews Drug discovery. 2021;20(2):101-124.
7.    Cao Z, Zuo X, Liu X, Xu G, Yong K-T. Recent progress in stimuli-responsive polymeric micelles for targeted delivery of functional nanoparticles. Adv Colloid Interface Sci. 2024;330:103206.
8.    Gomte SS, Agnihotri TG, Khopade S, Jain A. Exploring the potential of pH-sensitive polymers in targeted drug delivery. Journal of Biomaterials Science, Polymer Edition. 2023;35(2):228-268.
9.    Park H, Saravanakumar G, Kim J, Lim J, Kim WJ. Tumor Microenvironment Sensitive Nanocarriers for Bioimaging and Therapeutics. Advanced Healthcare Materials. 2020;10(5).
10.    Xu M, Qian J, Suo A, Wang H, Yong X, Liu X, et al. Reduction/pH dual-sensitive PEGylated hyaluronan nanoparticles for targeted doxorubicin delivery. Carbohydr Polym. 2013;98(1):181-188.
11.    Ghosh B, Biswas S. Polymeric micelles in cancer therapy: State of the art. J Controlled Release. 2021;332:127-147.
12.    Sharifi-Rad J, Quispe C, Patra JK, Singh YD, Panda MK, Das G, et al. Paclitaxel: Application in Modern Oncology and Nanomedicine-Based Cancer Therapy. Oxidative medicine and cellular longevity. 2021;2021:3687700-3687700.
13.    Recent Advances in Paclitaxel Drug Delivery: Challenges, Innovations, and Future Directions. Journal of Angiotherapy. 2024;8(8):1-12.
14.    Wang X, Gigant B, Zheng X, Chen Q. Microtubule‐targeting agents for cancer treatment: Seven binding sites and three strategies. MedComm – Oncology. 2023;2(3).
15.    Smith ER, Chen Z-S, Xu X-X. Paclitaxel and cancer treatment: Non-mitotic mechanisms of paclitaxel action in cancer therapy. Paclitaxel: Elsevier; 2022. p. 269-286.
16.    Yukuyama MN, de Souza A, Henostroza MAB, de Araujo GLB, Löbenberg R, Faria RdO, et al. Unveiling microtubule dynamics in lung cancer: Recent findings and prospects for drug delivery and treatment. J Drug Deliv Sci Technol. 2023;89:105017.
17.    Bai Z, Zhou Y, Peng Y, Ye X, Ma L. Perspectives and mechanisms for targeting mitotic catastrophe in cancer treatment. Biochim Biophys Acta. 2023;1878(5):188965.
18.    Neophytou CM, Panagi M, Stylianopoulos T, Papageorgis P. The Role of Tumor Microenvironment in Cancer Metastasis: Molecular Mechanisms and Therapeutic Opportunities. Cancers. 2021;13(9):2053.
19.    Singla AK, Garg A, Aggarwal D. Paclitaxel and its formulations. Int J Pharm. 2002;235(1-2):179-192.
20.    Ingole S, Vasdev N, Tekade M, Gupta T, Pawar B, Mhatre M, et al. Toxic effects of cancer therapies. Public Health and Toxicology Issues Drug Research, Volume 2: Elsevier; 2024. p. 353-379.
21.    Alalawy AI. Key genes and molecular mechanisms related to Paclitaxel Resistance. Cancer cell international. 2024;24(1):244-244.
22.    Karimi M, Eslami M, Sahandi-Zangabad P, Mirab F, Farajisafiloo N, Shafaei Z, et al. pH-Sensitive stimulus-responsive nanocarriers for targeted delivery of therapeutic agents. Wiley interdisciplinary reviews Nanomedicine and nanobiotechnology. 2016;8(5):696-716.
23.    Imran M, Shah MR, Shafiullah. Amphiphilic block copolymers–based micelles for drug delivery. Design and Development of New Nanocarriers: Elsevier; 2018. p. 365-400.
24.    Gaucher G, Dufresne M-H, Sant VP, Kang N, Maysinger D, Leroux J-C. Block copolymer micelles: preparation, characterization and application in drug delivery. J Controlled Release. 2005;109(1-3):169-188.
25.    Kalinova R, Dimitrov I. Functional Polyion Complex Micelles for Potential Targeted Hydrophobic Drug Delivery. Molecules (Basel, Switzerland). 2022;27(7):2178.
26.    Bami MS, Raeisi Estabragh MA, Khazaeli P, Ohadi M, Dehghannoudeh G. pH-responsive drug delivery systems as intelligent carriers for targeted drug therapy: Brief history, properties, synthesis, mechanism and application. J Drug Deliv Sci Technol. 2022;70:102987.
27.    Athulya KR, Varughese T, Kumar AC. Polymersomes: Beyond Basics‐Synthesis, Stimuli Response and Biomedical Applications. ChemistrySelect. 2024;9(36).
28.    Jia F, Su S, Zhang R, Wu Y. Main Fabrication Methods of Micellar Nanoparticles for Nanoscale Tumor Therapy through the Self-assembly of Amphiphilic Copolymers. Current Chinese Science. 2022;2(4):263-274.
29.    Kuperkar K, Patel D, Atanase LI, Bahadur P. Amphiphilic Block Copolymers: Their Structures, and Self-Assembly to Polymeric Micelles and Polymersomes as Drug Delivery Vehicles. Polymers. 2022;14(21):4702.
30.    Wang L, Cai F, Li Y, Lin X, Wang Y, Liang W, et al. pH-Responsive Block Copolymer Micelles of Temsirolimus: Preparation, Characterization and Antitumor Activity Evaluation. International journal of nanomedicine. 2024;19:9821-9841.
31.    Wang Z, Deng X, Ding J, Zhou W, Zheng X, Tang G. Mechanisms of drug release in pH-sensitive micelles for tumour targeted drug delivery system: A review. Int J Pharm. 2018;535(1-2):253-260.
32.    Chen Z, Wang X, Zhao N, Chen H, Guo G. Advancements in pH-responsive nanocarriers: enhancing drug delivery for tumor therapy. Expert Opinion on Drug Delivery. 2023;20(11):1623-1642.
33.    Zhuo S, Zhang F, Yu J, Zhang X, Yang G, Liu X. pH-Sensitive Biomaterials for Drug Delivery. Molecules (Basel, Switzerland). 2020;25(23):5649.
34.    pH-Responsive Polymer Nanoparticles. Advanced Polymer Nanoparticles: CRC Press; 2010. p. 181-208.
35.    AlSawaftah NM, Awad NS, Pitt WG, Husseini GA. pH-Responsive Nanocarriers in Cancer Therapy. Polymers. 2022;14(5):936.
36.    Zi Y, Yang K, He J, Wu Z, Liu J, Zhang W. Strategies to enhance drug delivery to solid tumors by harnessing the EPR effects and alternative targeting mechanisms. Adv Drug Del Rev. 2022;188:114449.
37.    Sun Y, Davis E. Nanoplatforms for Targeted Stimuli-Responsive Drug Delivery: A Review of Platform Materials and Stimuli-Responsive Release and Targeting Mechanisms. Nanomaterials (Basel, Switzerland). 2021;11(3):746.
38.    Lv S, Tang Z, Zhang D, Song W, Li M, Lin J, et al. Well-defined polymer-drug conjugate engineered with redox and pH-sensitive release mechanism for efficient delivery of paclitaxel. J Controlled Release. 2014;194:220-227.
39.    Haddad R, Alrabadi N, Altaani B, Li T. Paclitaxel Drug Delivery Systems: Focus on Nanocrystals’ Surface Modifications. Polymers. 2022;14(4):658.
40.    Gannimani R, Walvekar P, Naidu VR, Aminabhavi TM, Govender T. Acetal containing polymers as pH-responsive nano-drug delivery systems. J Controlled Release. 2020;328:736-761.
41.    Kubiak T. Polymeric capsules and micelles as promising carriers of anticancer drugs. Polymers in Medicine. 2022;52(1):37-50.
42.    Ezrahi S, Aserin A, Garti N. Basic principles of drug delivery systems – the case of paclitaxel. Adv Colloid Interface Sci. 2019;263:95-130.
43.    Yadav AS, Kundu IG, Radharani NNV, Mishra B, Kundu GC. pH-responsive nanomedicine for breast cancer targeting. Targeted Nanomedicine for Breast Cancer Therapy: Elsevier; 2022. p. 335-349.
44.    Tan RYH, Lee CS, Pichika MR, Cheng SF, Lam KY. PH Responsive Polyurethane for the Advancement of Biomedical and Drug Delivery. Polymers. 2022;14(9):1672.
45.    Sia CS, Lim HP, Tey BT, Goh B-H, Low LE. Stimuli-responsive nanoassemblies for targeted delivery against tumor and its microenvironment. Biochim Biophys Acta. 2022;1877(5):188779.
46.    Qiu N, Du X, Ji J, Zhai G. A review of stimuli-responsive polymeric micelles for tumor-targeted delivery of curcumin. Drug Dev Ind Pharm. 2021;47(6):839-856.
47.    Kumar S, Arora A, Pant V, Guchhait S, Kumar R, Mathur D, et al. Advances in Drug Delivery Systems for Lipophilic Drug Paclitaxel: Developments, Challenges, and Opportunities (A Review).  uss J Bioorg Chem. 2024;50(5):1752-1782.
48.    Wang Q, Atluri K, Tiwari AK, Babu RJ. Exploring the Application of Micellar Drug Delivery Systems in Cancer Nanomedicine. Pharmaceuticals (Basel, Switzerland). 2023;16(3):433.
49.    Liu J, Huang Y, Kumar A, Tan A, Jin S, Mozhi A, et al. pH-Sensitive nano-systems for drug delivery in cancer therapy. Biotechnol Adv. 2014;32(4):693-710.
50.    Danhier F, Danhier P, De Saedeleer CJ, Fruytier A-C, Schleich N, Rieux Ad, et al. Paclitaxel-loaded micelles enhance transvascular permeability and retention of nanomedicines in tumors. Int J Pharm. 2015;479(2):399-407.
51.    Jiang L, Li L, He X, Yi Q, He B, Cao J, et al. Overcoming drug-resistant lung cancer by paclitaxel loaded dual-functional liposomes with mitochondria targeting and pH-response. Biomaterials. 2015;52:126-139.
52.    Turner AP, Alam C, Bendayan R. Efflux transporters in cancer resistance: Molecular and functional characterization of P-glycoprotein. Drug Efflux Pumps in Cancer Resistance Pathways: From Molecular Recognition and Characterization to Possible Inhibition Strategies in Chemotherapy: Elsevier; 2020. p. 1-30.
53.    Jiang Y, Zhou Y, Zhang CY, Fang T. Co-Delivery of Paclitaxel and Doxorubicin by pH-Responsive Prodrug Micelles for Cancer Therapy. International journal of nanomedicine. 2020;15:3319-3331.
54.    Han Y, Pan J, Liang N, Gong X, Sun S. A pH-Sensitive Polymeric Micellar System Based on Chitosan Derivative for Efficient Delivery of Paclitaxel. International journal of molecular sciences. 2021;22(13):6659.
55.    Qi P, Bu Y, Xu J, Qin B, Luan S, Song S. pH-responsive release of paclitaxel from hydrazone-containing biodegradable micelles. Colloid Polym Sci. 2016;295(1):1-12.
56.    Xiang J, Wu B, Zhou Z, Hu S, Piao Y, Zhou Q, et al. Synthesis and evaluation of a paclitaxel-binding polymeric micelle for efficient breast cancer therapy. Science China Life Sciences. 2018;61(4):436-447.