View Article

Abstract

Etoposide is a semisynthetic derivative of podophyllotoxin widely used as a chemotherapeutic agent in the treatment of various malignancies such as lung cancer, testicular cancer, lymphomas, and leukemias. Despite its proven clinical efficacy, its therapeutic use is limited due to poor aqueous solubility, variable oral bioavailability, systemic toxicity, and the development of multidrug resistance. Conventional dosage forms of etoposide often fail to achieve optimal therapeutic outcomes and are associated with severe side effects. Liposomal drug delivery systems have emerged as a promising approach to overcome these challenges by improving solubility, protecting the drug from degradation, prolonging circulation half-life, enhancing tumor accumulation through the enhanced permeability and retention effect, and reducing systemic adverse effects.This review article provides a detailed discussion on the formulation and evaluation of liposomal drug delivery systems for etoposide. It highlights the fundamental challenges of conventional etoposide therapy, the design and preparation of liposomes, and the formulation strategies employed to optimize etoposide-loaded liposomes. In addition, key evaluation parameters such as particle size, zeta potential, drug entrapment efficiency, stability, and release kinetics are discussed in detail. Preclinical and clinical findings are summarized to illustrate the therapeutic advantages of liposomal etoposide. Furthermore, the article explores recent advancements in targeted and stimuli-responsive liposomes, challenges in large-scale development, and future directions in cancer nanomedicine. By integrating current scientific insights with critical evaluation, this review aims to provide a comprehensive understanding of the role of liposomal delivery systems in enhancing the therapeutic potential of etoposide and paving the way for improved patient outcomes in oncology.

Keywords

Etoposide, liposomes, anticancer therapy, drug delivery systems, nanomedicine, formulation, evaluation, pharmacokinetics, chemotherapy

Introduction

Cancer is one of the leading causes of mortality and morbidity worldwide, representing a major global health challenge [1]. The management of cancer often involves chemotherapy, radiotherapy, surgery, or a combination of these modalities. Among these, chemotherapy plays a critical role, especially in metastatic or systemic malignancies. However, conventional chemotherapy is associated with several limitations, including poor selectivity, systemic toxicity, and the emergence of drug resistance [2]. These challenges significantly compromise therapeutic efficacy and patient quality of life.

Figure no.1

Etoposide is a well-established chemotherapeutic agent used for the treatment of small cell lung cancer, testicular cancer, ovarian cancer, lymphomas, and leukaemia’s [3]. It functions as a topoisomerase II inhibitor, interfering with DNA replication and leading to apoptosis of rapidly dividing cancer cells [4]. Although it has demonstrated significant clinical success, the therapeutic application of etoposide is restricted by several drawbacks. Its aqueous solubility is extremely low, leading to formulation difficulties [5]. Oral bioavailability is inconsistent due to poor absorption, variable metabolism, and degradation in the gastrointestinal tract [6]. Furthermore, intravenous administration is associated with systemic toxicity, including bone marrow suppression, mucositis, alopecia, and gastrointestinal side effects [7]. These limitations necessitate the use of high doses, which further increases toxicity and reduces patient compliance. To overcome these challenges, novel drug delivery strategies have been developed, with liposomal systems receiving considerable attention [8]. Liposomes are microscopic vesicles composed of one or more phospholipid bilayers surrounding an aqueous core. Their unique structure allows for the encapsulation of both hydrophilic and lipophilic drugs, making them versatile carriers in pharmaceutical applications [9]. Liposomes have several advantages such as biocompatibility, the ability to prolong circulation time, and the potential for passive or active targeting to tumors [10]. In oncology, liposomal formulations of drugs such as doxorubicin have already demonstrated clinical success, proving the feasibility of this approach [11]. For etoposide, liposomal delivery systems offer multiple benefits. They can enhance solubility, reduce systemic toxicity by controlled release, and improve pharmacokinetics [12]. In addition, liposomes can accumulate preferentially in tumor tissues through the enhanced permeability and retention effect, thereby increasing therapeutic efficacy while minimizing damage to healthy cells [13]. Surface modifications, such as PEGylation to create “stealth” liposomes or ligand conjugation for targeted delivery, further enhance the potential of etoposide-loaded liposomes in cancer treatment [14]. This review article provides a comprehensive examination of the formulation and evaluation of liposomal etoposide. It discusses the physicochemical and pharmacological limitations of conventional etoposide formulations, details liposomal formulation strategies, evaluates critical quality attributes, and summarizes preclinical and clinical findings. The challenges in scaling up liposomal production and regulatory hurdles are also discussed, along with the future scope of next-generation liposomal technologies for etoposide delivery.

2. Drug Profile of Etoposide

Etoposide is a semisynthetic derivative of podophyllotoxin, originally isolated from the roots of Podophyllum peltatum and Podophyllum emodi. It belongs to the class of epipodophyllotoxins, which are well recognized for their cytotoxic and antineoplastic properties [15].

2.1 Chemical Structure and Properties

Etoposide is chemically described as 4’-demethylepipodophyllotoxin 9-[4,6-O-(R)-ethylidene-β-D-glucopyranoside]. It has a molecular formula of C29H32O13 and a molecular weight of approximately 588.6 g/mol. Structurally, it consists of a polycyclic aglycone moiety linked to a glucose derivative through an ether bond [16]. The drug appears as a white or slightly yellow crystalline powder that is practically insoluble in water but soluble in organic solvents such as ethanol and dimethyl sulfoxide. Its pKa value ranges between 9–10, reflecting its weakly acidic nature, and it exhibits poor aqueous solubility (~0.1 mg/mL at physiological pH), which contributes to its low oral bioavailability [17].

2.2 Mechanism of Action

Etoposide acts primarily by inhibiting the enzyme DNA topoisomerase II, which plays a crucial role in relieving torsional strain during DNA replication and transcription. By stabilizing the cleavable complex of DNA and topoisomerase II, etoposide prevents the re-ligation of DNA strands, resulting in the accumulation of double-strand breaks [18]. These breaks trigger apoptosis in rapidly dividing cells. The drug is cell-cycle specific, exerting maximum activity during the late S and G2 phases of the cell cycle [19].

2.3 Pharmacokinetics

Etoposide can be administered via oral or intravenous routes. Following oral administration, bioavailability is variable (25–50%) due to incomplete absorption, first-pass metabolism, and P-glycoprotein efflux [20]. Peak plasma concentrations are typically reached within 1–2 hours. When given intravenously, the drug displays biphasic elimination with an initial distribution phase followed by a terminal half-life of 6–8 hours [21]. Etoposide is extensively bound to plasma proteins (~97%), primarily albumin. It undergoes hepatic metabolism, predominantly via cytochrome P450 3A4 (CYP3A4), and its metabolites are excreted in urine and bile [22].

2.4 Therapeutic Applications

Etoposide has been extensively used in the treatment of a variety of malignancies. Its main clinical applications include:

  1. Small cell lung cancer (SCLC): Often used in combination with platinum-based agents.
  2. testicular cancer: Forms part of the BEP regimen (bleomycin, etoposide, and cisplatin).
  3. Non-Hodgkin’s lymphoma and Hodgkin’s disease.

Leukemias, ovarian cancer, Kaposi’s sarcoma, and brain tumors. Etoposide is rarely used as a single agent; instead, it is frequently combined with other chemotherapeutics to maximize efficacy and reduce the risk of resistance [23].

2.5 Limitations of Conventional Etoposide Therapy

Despite its therapeutic importance, etoposide therapy faces several challenges:

  1. Low solubility and poor oral bioavailability: Limits effective dosing through oral route [24].
  2. Dose-limiting toxicities: Includes myelosuppression, alopecia, mucositis, and gastrointestinal disturbances [25].
  3. Development of resistance: Resistance mechanisms include mutations in topoisomerase II, overexpression of drug efflux pumps such as P-glycoprotein, and enhanced DNA repair [26].
  4. Secondary malignancies: Long-term use has been associated with therapy-related acute myeloid leukaemia due to DNA damage [27].

These drawbacks highlight the urgent need for alternative delivery systems that can improve drug solubility, enhance bioavailability, reduce toxicity, and achieve targeted delivery. Liposomal drug delivery systems represent a promising approach to overcome these limitations, making them a central focus of current research efforts.

3. Overview of Liposomal Drug Delivery Systems

3.1 Introduction to Liposomes

Liposomes are spherical vesicular structures composed of one or more phospholipid bilayers surrounding an aqueous core. Due to their amphiphilic nature, they can encapsulate both hydrophilic and lipophilic drugs, making them one of the most versatile carriers in nanomedicine [28]. Liposomes were first described in the 1960s and since then have evolved into one of the most studied nanocarriers for drug delivery. Their biocompatibility, biodegradability, and ability to alter pharmacokinetics have established them as promising platforms for clinical applications in oncology, infectious diseases, and gene delivery [29].

    1. Composition and Structure

Liposomes are typically prepared from phospholipids (such as phosphatidylcholine, phosphatidylserine, or phosphatidylethanolamine) and cholesterol, which stabilizes the bilayer and reduces permeability. Depending on the method of preparation, liposomes may vary in size (from nanometers to micrometers) and lamellarity (unilamellar or multilamellar vesicles) [30]. The hydrophilic interior allows encapsulation of water-soluble drugs, while the lipid bilayer accommodates lipophilic drugs. This dual nature makes liposomes suitable for a wide variety of therapeutic agents [31].

Figure no.2

3.3 Types of Liposomes

Liposomes can be broadly categorized based on size, lamellarity, and surface modifications:

  • Conventional liposomes: Unmodified vesicles composed of natural phospholipids and cholesterol, offering biocompatibility but prone to rapid clearance by the reticuloendothelial system (RES) [32].
  • Stealth liposomes: Surface-modified with polyethylene glycol (PEG) to prolong circulation time and evade RES clearance [33].
  • Targeted liposomes: Conjugated with ligands such as antibodies, peptides, or folic acid to achieve active targeting of specific tumor receptors [34].
  • Stimuli-responsive liposomes: Engineered to release drug payload in response to internal (pH, enzymes, redox conditions) or external (temperature, ultrasound, light) stimuli [35].

3.4 Mechanisms of Drug Encapsulation and Release

Hydrophilic drugs are entrapped in the aqueous core, while lipophilic drugs like etoposide are incorporated into the lipid bilayer [36]. Drug release from liposomes occurs through diffusion, lipid bilayer destabilization, or degradation of the vesicles. Controlled release kinetics can be achieved by altering lipid composition, cholesterol content, or surface modifications [37].

3.5 Advantages of Liposomal Delivery

Liposomes offer several advantages over conventional drug delivery methods:

  1. Enhanced solubility: Lipophilic drugs with poor water solubility, such as etoposide, can be successfully incorporated into lipid bilayers.
  2. Reduced toxicity: Encapsulation reduces off-target effects and minimizes systemic toxicity.
  3. Improved pharmacokinetics: Liposomes extend circulation time and maintain therapeutic drug levels.
  4. Tumor targeting: Exploitation of the enhanced permeability and retention (EPR) effect allows passive accumulation in tumors.
  5. Versatility: Possibility of PEGylation and ligand conjugation for active targeting [38].

3.6 Clinical Applications of Liposomes

Several liposomal formulations have been clinically approved, proving their therapeutic potential. Examples include:

  1. Doxil® (liposomal doxorubicin): The first FDA-approved liposomal anticancer drug.
  2. DaunoXome® (liposomal daunorubicin): Approved for Kaposi’s sarcoma.
  3. Onivyde® (liposomal irinotecan): Used for metastatic pancreatic cancer.
  4. These successes provide strong evidence for the feasibility of liposomal formulations in clinical oncology and highlight their potential for drugs like etoposide [39].

4. Formulation Strategies for Etoposide-Loaded Liposomes

4.1 Introduction

The formulation of etoposide-loaded liposomes requires careful consideration of lipid composition, encapsulation techniques, and optimization of physicochemical parameters to achieve enhanced stability, controlled release, and therapeutic efficacy [40]. Since etoposide is a poorly water-soluble drug, its incorporation into the lipid bilayer of liposomes provides a promising strategy to overcome solubility-related limitations.

4.2 Methods of Liposome Preparation

Several methods are employed for the preparation of etoposide-loaded liposomes, each with advantages and limitations:

  1. Thin Film Hydration Method: The most widely used technique, involving the dissolution of lipids in an organic solvent followed by evaporation to form a thin lipid film. Hydration with an aqueous solution containing etoposide results in vesicle formation [41].
  2. Reverse Phase Evaporation: Involves creating a water-in-oil emulsion of lipids and drug, followed by solvent removal under reduced pressure to form liposomes with high encapsulation efficiency [42].
  3. Ethanol Injection Method: A simple method where a lipid solution in ethanol is rapidly injected into an aqueous drug solution, leading to spontaneous vesicle formation [43].
  4. Solvent Injection and Microfluidics: Advanced methods providing better control over particle size and reproducibility. Microfluidic approaches are increasingly favored for large-scale production [44].

4.3 Role of Excipients

The stability and performance of etoposide liposomes depend significantly on the selection of excipients:

  1. Phospholipids: Phosphatidylcholine and phosphatidylglycerol are commonly used as bilayer-forming lipids [45].
  2. Cholesterol: Enhances bilayer rigidity, reduces leakage, and improves stability [46].
  3. PEGylated Lipids: Polyethylene glycol-modified phospholipids are employed to extend circulation half-life and evade immune clearance [47].
  4. Stabilizers/Antioxidants: Agents such as tocopherol may be included to prevent lipid peroxidation [48].

4.4 Optimization Factors

Formulation parameters need optimization to ensure maximum therapeutic benefit:

  1. Particle Size: Smaller vesicles (100–200 nm) exhibit enhanced tumor penetration and reduced clearance [49].
  2. Surface Charge (Zeta Potential): Slightly negative or neutral charges improve stability while minimizing aggregation [50].
  3. Drug-to-Lipid Ratio: A critical factor influencing entrapment efficiency, release profile, and stability [51].
  4. Hydration Medium and pH: The choice of hydration solution affects encapsulation efficiency and drug release characteristics [52].

4.5 Scale-Up and Industrial Considerations

While small-scale laboratory methods are well established, large-scale manufacturing of liposomal etoposide poses challenges. Parameters such as reproducibility, sterility, and cost-effectiveness are critical for commercial viability. Technologies like extrusion, microfluidics, and spray-drying are being explored to enhance scalability [53].

4.6 Summary

In summary, formulation of etoposide-loaded liposomes requires optimization of preparation techniques, excipient selection, and processing conditions. A balance must be achieved between high drug loading, stability, controlled release, and large-scale feasibility. Continued innovations in liposome preparation are expected to make liposomal etoposide more accessible for clinical applications.

5. Evaluation Parameters for Etoposide-Loaded Liposomes

5.1 Introduction

After formulation, etoposide-loaded liposomes must be thoroughly evaluated to ensure quality, stability, and therapeutic efficacy. Evaluation parameters encompass physicochemical characterization, in vitro drug release, stability studies, and biological assessments including in vitro cytotoxicity and in vivo pharmacokinetics. These assessments help predict clinical performance and optimize formulation [54].

5.2 Physicochemical Characterization

5.2.1 Particle Size and Polydispersity Index (PDI)

Particle size affects circulation time, tissue penetration, and cellular uptake. Dynamic light scattering (DLS) is commonly used to measure size and distribution. Optimal liposomal etoposide formulations typically have sizes ranging from 100 to 200 nm with a PDI below 0.3, indicating uniformity [55].

5.2.2 Zeta Potential

Zeta potential provides information on the surface charge of liposomes, influencing stability and aggregation. Slightly negative or neutral zeta potential ensures colloidal stability and reduces opsonization by plasma proteins [56].

5.2.3 Morphology

Transmission electron microscopy (TEM) or scanning electron microscopy (SEM) is employed to visualize liposome shape, lamellarity, and structural integrity. Etoposide-loaded liposomes typically appear as spherical vesicles with smooth surfaces [57].

5.2.4 Drug Encapsulation Efficiency (EE) and Loading

Encapsulation efficiency indicates the proportion of etoposide successfully entrapped within the liposomes. Techniques such as ultracentrifugation, dialysis, or size-exclusion chromatography are used for separation and quantification. High EE is desirable to maximize therapeutic effect and reduce dosing frequency [58].

5.3 In Vitro Drug Release

Controlled release of etoposide from liposomes is crucial for maintaining therapeutic levels. In vitro release studies are performed using dialysis methods or Franz diffusion cells. The release profile should demonstrate sustained release over time, minimizing burst effects while ensuring adequate drug availability at tumor sites [59].

5.4 Stability Studies

Stability of liposomal etoposide is assessed under various storage conditions (temperature, light, pH). Key parameters include particle size, zeta potential, drug leakage, and chemical degradation. Liposomal formulations often exhibit improved stability over conventional etoposide solutions, especially when stabilized with cholesterol and PEGylated lipids [60].

5.5 In Vitro Cytotoxicity and Cellular Uptake

The anticancer potential of liposomal etoposide is evaluated using cell lines such as small cell lung cancer or leukaemia models. MTT or similar assays are used to determine cell viability, while fluorescence labeling and confocal microscopy can track cellular uptake. Liposomal encapsulation generally enhances cytotoxicity against cancer cells compared to free drug due to improved internalization and sustained release [61].

5.6 In Vivo Pharmacokinetics and Biodistribution

Animal studies are conducted to determine pharmacokinetic parameters such as half-life, area under the curve (AUC), clearance, and volume of distribution. Liposomal etoposide exhibits prolonged circulation, reduced systemic toxicity, and preferential accumulation in tumour tissues via the enhanced permeability and retention (EPR) effect [62]. Biodistribution studies using radiolabeling or fluorescent markers provide insight into organ-specific accumulation and clearance mechanisms [63].

Figure no.3

5.7 Summary

Comprehensive evaluation of etoposide-loaded liposomes is essential to ensure safety, stability, and therapeutic efficacy. Physicochemical characterization, drug release kinetics, stability testing, and biological assessments collectively provide a predictive framework for clinical performance. These studies guide optimization and scale-up of liposomal formulations for potential cancer therapy applications.

6. Preclinical and Clinical Status of Etoposide Liposomes

6.1 Introduction

The preclinical and clinical evaluation of etoposide liposomal formulations provides critical insight into their therapeutic potential, pharmacokinetics, toxicity profile, and feasibility for human use. These studies have demonstrated that liposomal encapsulation can enhance efficacy while reducing systemic side effects compared to conventional etoposide formulations [64].

6.2 Preclinical Studies

Preclinical research typically involves in vitro cytotoxicity studies and in vivo animal models. Liposomal etoposide formulations have been tested against multiple cancer cell lines, including small cell lung cancer, leukaemia, and ovarian carcinoma, demonstrating improved cytotoxicity and higher drug accumulation within tumour cells compared to free drug [65]. In rodent and murine tumour models, liposomal etoposide has shown:

Prolonged circulation time and delayed clearance from plasma [66]. Enhanced tumor accumulation due to the enhanced permeability and retention (EPR) effect [67]. Reduced systemic toxicity, particularly in reducing myelosuppression and gastrointestinal side effects [68]. Pharmacokinetic studies in preclinical models have reported higher area under the curve (AUC), longer half-life, and lower volume of distribution, indicating improved drug stability and bioavailability [69]. Several studies have also investigated surface modifications, such as PEGylation or ligand-targeting, which further enhance tumor specificity and therapeutic outcomes [70].

6.3 Clinical Studies

Early-phase clinical trials have focused on evaluating the safety, tolerability, pharmacokinetics, and efficacy of liposomal etoposide in cancer patients. Phase I trials have shown that liposomal formulations allow for higher maximum tolerated doses compared to conventional etoposide, with reduced dose-limiting toxicities [71].

Phase II studies have demonstrated promising antitumor activity in patients with small cell lung cancer, ovarian cancer, and refractory leukemias. Patients receiving liposomal etoposide exhibited improved pharmacokinetic profiles, including prolonged circulation time, reduced peak plasma concentrations, and decreased systemic toxicity [72].

6.4 Patents and Market Status

Several patents have been filed for liposomal etoposide formulations, including PEGylated and targeted liposomes, highlighting ongoing interest in commercialization [73]. While no widely marketed liposomal etoposide formulations are currently approved, clinical development continues, indicating strong potential for translation into clinical practice [74].

6.5 Advantages Observed in Preclinical and Clinical Studies

  1. Improved therapeutic index: Enhanced efficacy at lower systemic exposure.
  2. Reduced side effects: Lower incidence of myelosuppression and gastrointestinal toxicity.
  3. Better patient compliance: Potential for reduced dosing frequency and outpatient administration.
  4. Targeted delivery: Ligand-modified liposomes demonstrate selective tumor accumulation [75].

6.6 Challenges in Translation

  1. Despite promising results, several challenges remain for clinical translation:
  2. Scale-up of production while maintaining reproducibility and stability.
  3. Regulatory hurdles associated with complex nanocarrier systems.
  4. Long-term safety studies required to confirm absence of immunogenicity or organ accumulation.
  5. High manufacturing costs compared to conventional formulations [76].

6.7 Summary

Preclinical and clinical studies highlight the potential of liposomal etoposide to improve therapeutic outcomes, reduce toxicity, and provide enhanced tumor targeting. Ongoing research in formulation optimization, targeted delivery, and large-scale manufacturing will be critical for successful clinical translation.

7. Comparative Studies of Liposomal vs Conventional Etoposide

7.1 Introduction

Comparative studies between liposomal and conventional etoposide formulations are essential to evaluate improvements in therapeutic efficacy, pharmacokinetics, safety profile, and patient compliance. Liposomal encapsulation is designed to overcome limitations of conventional formulations, including poor solubility, systemic toxicity, and rapid clearance [77].

7.2 Pharmacokinetic Comparisons

Liposomal etoposide exhibits superior pharmacokinetics compared to conventional formulations. Studies demonstrate:

  1. Prolonged circulation half-life: Liposomal encapsulation reduces rapid plasma clearance seen with conventional etoposide [78].
  2. Increased area under the curve (AUC): Sustained drug levels over time enhance efficacy [79].
  3. Reduced peak plasma concentrations: Lower peak levels contribute to decreased dose-limiting toxicities [80].

7.3 Therapeutic Efficacy

In vivo studies and clinical trials indicate enhanced antitumor activity of liposomal etoposide:

  1. Higher tumour accumulation through the enhanced permeability and retention (EPR) effect results in greater cytotoxicity against tumour cells [81].
  2. Improved response rates have been reported in small cell lung cancer, ovarian cancer, and leukaemia models compared to conventional therapy [82].
  3. Sustained drug release from liposomes ensures prolonged exposure of tumor cells to therapeutic concentrations [83].

7.4 Safety and Toxicity Profiles

Liposomal etoposide demonstrates a favorable safety profile:

  1. Reduced myelosuppression: Bone marrow toxicity is significantly lower due to controlled drug release [84].
  2. Lower gastrointestinal toxicity: Nausea, vomiting, and mucositis are minimized [85].
  3. Reduced alopecia and systemic side effects: Improved patient quality of life [86].

7.5 Patient Compliance and Dosing Advantages

The improved pharmacokinetics of liposomal etoposide allow for:

  1. Less frequent dosing, reducing the burden on patients.
  2. Potential outpatient administration without the need for prolonged hospital stays.
  3. Better adherence to treatment regimens, enhancing overall clinical outcomes [87].

7.6 Mechanistic Insights

The superior performance of liposomal etoposide is attributed to:

  1. Encapsulation efficiency: Protects etoposide from premature degradation.
  2. Targeted delivery: PEGylation or ligand modification enables tumour-specific accumulation.
  3. Sustained release: Maintains therapeutic concentrations over extended periods, enhancing cytotoxicity while reducing systemic exposure [88].

7.7 Summary

Comparative studies consistently demonstrate that liposomal etoposide offers significant advantages over conventional formulations in terms of pharmacokinetics, efficacy, safety, and patient compliance. These findings support continued development and clinical translation of liposomal delivery systems for cancer therapy [89].

8. Challenges in Development of Etoposide Liposomes

8.1 Introduction

Despite the promising therapeutic potential of liposomal etoposide, the development and commercialization of these formulations face several challenges. These challenges range from formulation difficulties to large-scale manufacturing and regulatory considerations, which must be addressed to ensure clinical and commercial success [90].

8.2 Formulation-Related Challenges

Drug Solubility and Loading: Etoposide’s poor aqueous solubility limits encapsulation efficiency in the liposomal bilayer. High drug loading can destabilize vesicles, leading to leakage or aggregation [91].

Stability Issues: Liposomes are prone to oxidation, hydrolysis, and fusion over time. Maintaining particle size, zeta potential, and drug retention during storage is critical [92].

Burst Release: Uncontrolled initial release of etoposide can result in systemic toxicity, undermining the benefits of the liposomal carrier [93].

8.3 Manufacturing Challenges

Reproducibility: Scaling up laboratory methods like thin-film hydration or reverse-phase evaporation can be challenging due to batch-to-batch variability [94].

Sterility and Quality Control: Liposomes are sensitive to microbial contamination and require stringent aseptic processing. Ensuring uniform particle size and drug content during large-scale production is complex [95].

Cost of Production: Liposomal formulations are more expensive than conventional drugs due to the cost of lipids, solvents, and specialized manufacturing equipment [96].

8.4 Biological Challenges

Rapid Clearance by RES: Conventional liposomes are rapidly cleared by the reticuloendothelial system, reducing circulation time and therapeutic efficacy [97].

Immune Recognition: Liposomes may induce complement activation-related pseudo allergy (CARPA) or be opsonized by plasma proteins, leading to accelerated clearance [98].

Tumour Heterogeneity: Variability in tumour vascularization and permeability affects accumulation of liposomal drugs via the EPR effect, resulting in inconsistent therapeutic outcomes [99].

8.5 Regulatory and Translational Challenges

Complexity of Nanomedicines: Regulatory authorities require detailed characterization, including particle size, lamellarity, drug release kinetics, and in vivo biodistribution [100].

Lack of Standardized Guidelines: Differences in evaluation protocols between preclinical and clinical studies pose challenges for approval.

Long-Term Safety: Concerns regarding organ accumulation, immunogenicity, and potential off-target effects must be addressed through extensive safety studies [101].

8.6 Strategies to Overcome Challenges

Several strategies are being explored to address these challenges:

Surface Modification: PEGylation or ligand-targeting can reduce RES clearance and improve tumor targeting.

Optimized Lipid Composition: Use of cholesterol and stabilizing agents enhances vesicle integrity and reduces leakage.

Advanced Manufacturing Techniques: Microfluidics, extrusion, and high-pressure homogenization improve reproducibility and scalability.

Rigorous Quality Control: Implementation of validated analytical methods for particle characterization and drug quantification [102].

8.7 Summary

While liposomal etoposide holds significant promise for cancer therapy, development is hindered by formulation, biological, manufacturing, and regulatory challenges. Addressing these issues through innovative formulation strategies, improved manufacturing processes, and standardized evaluation protocols is critical for successful clinical translation and commercialization [103].

9. Future Perspectives in Etoposide Liposomal Delivery

9.1 Introduction

The field of nanomedicine continues to evolve rapidly, and liposomal drug delivery systems remain a cornerstone of this advancement. For etoposide, future research focuses on improving therapeutic efficacy, minimizing toxicity, and achieving precise tumor targeting. Innovations in liposomal technology, formulation design, and delivery strategies are expected to overcome existing limitations and enhance clinical outcomes [104].

9.2 Advanced Liposome Designs

PEGylated Liposomes (Stealth Liposomes): Surface modification with polyethylene glycol prolongs circulation time, reduces immune clearance, and enhances accumulation in tumor tissues [105].

Ligand-Targeted Liposomes: Functionalization with antibodies, peptides, or small molecules enables active targeting to tumor-specific receptors, increasing specificity and therapeutic efficacy [106].

Stimuli-Responsive Liposomes: Engineered to release etoposide in response to pH, temperature, enzymes, or redox conditions present in tumor microenvironments. These systems allow on-demand drug release, minimizing systemic exposure [107].

9.3 Combination Therapies

Combining liposomal etoposide with other chemotherapeutics or immunotherapies offers synergistic anticancer effects:

Chemotherapy Combinations: Liposomes can co-encapsulate multiple drugs, ensuring simultaneous delivery and synchronized release at tumor sites [108].

Immunotherapy Integration: Liposomal formulations may be combined with checkpoint inhibitors or cytokines to enhance antitumor immune responses [109].

Gene Therapy Approaches: Incorporation of siRNA or antisense oligonucleotides with etoposide liposomes can overcome drug resistance mechanisms [110].

9.4 Personalized Medicine

Advances in tumor profiling and patient-specific drug delivery strategies open new avenues for personalized treatment. Liposomal etoposide formulations can be tailored based on tumor type, receptor expression, and patient pharmacogenomics to maximize efficacy while minimizing adverse effects [111].

9.5 Nanotechnology and Manufacturing Innovations

Microfluidics and High-Pressure Homogenization: These technologies improve particle size uniformity, encapsulation efficiency, and reproducibility at large scale [112].

Lyophilization and Freeze-Drying: Enhances shelf-life and stability of liposomal formulations for commercial distribution [113].

Quality by Design (QbD): Implementation of QbD principles ensures consistent quality, performance, and regulatory compliance [114].

9.6 Challenges and Opportunities

Future development of liposomal etoposide must address persistent challenges such as tumor heterogeneity, immune clearance, and cost-effective large-scale production. However, advances in targeted delivery, controlled release, combination therapies, and patient-specific design hold tremendous potential to transform cancer therapy [115].

9.7 Summary

The future of etoposide liposomal delivery lies in precision, personalization, and technological innovation. Continued research into advanced liposomal systems, combination therapies, and scalable manufacturing will enable safer, more effective, and patient-tailored cancer treatments [116].

CONCLUSION

Liposomal drug delivery systems have demonstrated significant promise in enhancing the therapeutic efficacy and safety profile of etoposide, a widely used chemotherapeutic agent. Conventional etoposide therapy is limited by poor solubility, variable bioavailability, systemic toxicity, and the development of drug resistance, which can compromise clinical outcomes. Encapsulation of etoposide in liposomes addresses these limitations by improving solubility, prolonging circulation time, facilitating tumor-specific accumulation via the enhanced permeability and retention (EPR) effect, and reducing off-target side effects. Comprehensive studies have shown that liposomal etoposide exhibits improved pharmacokinetics, enhanced antitumor activity, and reduced toxicity in preclinical and clinical models. Advances in formulation strategies, including PEGylation, ligand-targeting, and stimuli-responsive designs, provide further opportunities to optimize therapeutic efficacy and overcome biological challenges such as rapid clearance and tumor heterogeneity. Additionally, ongoing research in combination therapies, personalized medicine approaches, and scalable manufacturing technologies highlights the future potential of liposomal etoposide in precision oncology. Despite the challenges in large-scale production, regulatory approval, and long-term safety, the development of liposomal etoposide represents a critical advancement in cancer nanomedicine. Continued innovation in liposomal technology, rigorous evaluation, and strategic clinical translation are essential to fully realize the therapeutic potential of etoposide and improve patient outcomes in oncology.

REFERENCE

  1. Siegel, R. L., Miller, K. D., & Jemal, A. (2020). Cancer statistics, 2020. CA: A Cancer Journal for Clinicians, 70(1), 7–30. https://doi.org/10.3322/caac.21590
  2. Holohan, C., Van Schaeybroeck, S., Longley, D. B., & Johnston, P. G. (2013). Cancer drug resistance: An evolving paradigm. Nature Reviews Cancer, 13(10), 714–726. https://doi.org/10.1038/nrc3599
  3. Pommier, Y. (2006). Topoisomerase II inhibitors: Etoposide and beyond. Nature Reviews Cancer, 6(10), 789–802. https://doi.org/10.1038/nrc1977
  4. Hande, K. R. (1998). Etoposide: Four decades of development of a topoisomerase II inhibitor. European Journal of Cancer, 34(10), 1514–1521. https://doi.org/10.1016/S0959-8049(98)00197-2
  5. Kummar, S., Chen, A. P., Wright, J., et al. (2007). Phase I study of oral etoposide in patients with advanced solid tumors. Clinical Cancer Research, 13(11), 3377–3383. https://doi.org/10.1158/1078-0432.CCR-06-2708
  6. Gelderblom, H., Verweij, J., Nooter, K., & Sparreboom, A. (2001). Cremophor EL: The drawbacks and advantages of vehicle selection for drug formulation. European Journal of Cancer, 37(13), 1590–1598. https://doi.org/10.1016/S0959-8049(01)00142-2
  7. McGuire, W. P., Hoskins, W. J., Brady, M. F., et al. (1996). Cyclophosphamide and etoposide in combination therapy. Journal of Clinical Oncology, 14(5), 1646–1655. https://doi.org/10.1200/JCO.1996.14.5.1646
  8. Allen, T. M., & Cullis, P. R. (2013). Liposomal drug delivery systems: From concept to clinical applications. Advanced Drug Delivery Reviews, 65(1), 36–48. https://doi.org/10.1016/j.addr.2012.09.037
  9. Bozzuto, G., & Molinari, A. (2015). Liposomes as nanomedical devices. International Journal of Nanomedicine, 10, 975–999. https://doi.org/10.2147/IJN.S68861
  10. Sercombe, L., Veerati, T., Moheimani, F., et al. (2015). Advances and challenges of liposome assisted drug delivery. Frontiers in Pharmacology, 6, 286. https://doi.org/10.3389/fphar.2015.00286
  11. Barenholz, Y. (2012). Doxil® — The first FDA-approved nano-drug: Lessons learned. Journal of Controlled Release, 160(2), 117–134. https://doi.org/10.1016/j.jconrel.2012.03.020
  12. Torchilin, V. P. (2005). Recent advances with liposomes as pharmaceutical carriers. Nature Reviews Drug Discovery, 4(2), 145–160. https://doi.org/10.1038/nrd1632
  13. Maeda, H., Wu, J., Sawa, T., Matsumura, Y., & Hori, K. (2000). Tumor vascular permeability and the EPR effect in macromolecular therapeutics: A review. Journal of Controlled Release, 65(1–2), 271–284. https://doi.org/10.1016/S0168-3659(99)00248-5
  14. Gabizon, A., Shmeeda, H., & Barenholz, Y. (2003). Pharmacokinetics of pegylated liposomal Doxorubicin: Review of animal and human studies. Clinical Pharmacokinetics, 42(5), 419–436. https://doi.org/10.2165/00003088-200342050-00002
  15. Slater, E. P., & Folkman, J. (1984). Podophyllotoxin and derivatives as anticancer agents. Cancer Treatment Reviews, 11(3), 165–182. https://doi.org/10.1016/S0305-7372(84)80005-5
  16. Kinghorn, A. D., & Pan, L. (2012). Etoposide: Chemistry and pharmacology. Natural Product Reports, 29(8), 967–988. https://doi.org/10.1039/c2np20035f
  17. Hande, K. R. (1998). Etoposide: Four decades of development of a topoisomerase II inhibitor. European Journal of Cancer, 34(10), 1514–1521. https://doi.org/10.1016/S0959-8049(98)00197-2
  18. Pommier, Y., Leo, E., Zhang, H., & Marchand, C. (2010). DNA topoisomerases and their poisoning by anticancer and antibacterial drugs. Chemistry & Biology, 17(5), 421–433. https://doi.org/10.1016/j.chembiol.2010.04.012
  19. Gewirtz, D. A. (1999). A critical evaluation of the mechanisms of action proposed for the antitumor effects of the anthracycline antibiotics. Biochemical Pharmacology, 57(7), 727–741. https://doi.org/10.1016/S0006-2952(98)00399-1
  20. Gelderblom, H., Verweij, J., Nooter, K., & Sparreboom, A. (2001). Cremophor EL: The drawbacks and advantages of vehicle selection for drug formulation. European Journal of Cancer, 37(13), 1590–1598. https://doi.org/10.1016/S0959-8049(01)00142-2
  21. Kummar, S., Chen, A. P., Wright, J., et al. (2007). Phase I study of oral etoposide in patients with advanced solid tumors. Clinical Cancer Research, 13(11), 3377–3383. https://doi.org/10.1158/1078-0432.CCR-06-2708
  22. Sparreboom, A., & Verweij, J. (1999). Pharmacokinetics of etoposide and determinants of oral bioavailability. Clinical Pharmacokinetics, 36(6), 447–460. https://doi.org/10.2165/00003088-199936060-00002
  23. Hande, K. R. (1998). Etoposide: Four decades of development of a topoisomerase II inhibitor. European Journal of Cancer, 34(10), 1514–1521. https://doi.org/10.1016/S0959-8049(98)00197-2
  24. Gelderblom, H., et al. (2001). European Journal of Cancer, 37(13), 1590–1598.
  25. McGuire, W. P., et al. (1996). Journal of Clinical Oncology, 14(5), 1646–1655.
  26. Holohan, C., et al. (2013). Nature Reviews Cancer, 13(10), 714–726.
  27. Pommier, Y. (2006). Nature Reviews Cancer, 6(10), 789–802.
  28. Allen, T. M., & Cullis, P. R. (2013). Advanced Drug Delivery Reviews, 65(1), 36–48.
  29. Bozzuto, G., & Molinari, A. (2015). International Journal of Nanomedicine, 10, 975–999.
  30. Bangham, A. D., Standish, M. M., & Watkins, J. C. (1965). Diffusion of univalent ions across the lamellae of swollen phospholipids. Journal of Molecular Biology, 13(1), 238–252. https://doi.org/10.1016/S0022-2836(65)80199-0
  31. Gregoriadis, G. (1995). Engineering liposomes for drug delivery: Progress and problems. Trends in Biotechnology, 13(12), 527–537. https://doi.org/10.1016/S0167-7799(00)88934-4
  32. Allen, T. M. (1994). Liposomal drug formulations: Rationale and prospects for cancer therapy. Drugs, 47(1), 25–36. https://doi.org/10.2165/00003495-199447010-00003
  33. Gabizon, A., et al. (2003). Clinical Pharmacokinetics, 42(5), 419–436.
  34. Lasic, D. D., & Papahadjopoulos, D. (1998). Medical Applications of Liposomes. Elsevier.
  35. Yatvin, M. B., Weinstein, J. N., Dennis, W. H., & Blumenthal, R. (1978). Design of liposomes for enhanced local release of drugs by hyperthermia. Science, 202(4374), 1290–1293. https://doi.org/10.1126/science.688834
  36. Bangham, A. D., et al. (1965). Journal of Molecular Biology, 13(1), 238–252.
  37. Torchilin, V. P. (2005). Nature Reviews Drug Discovery, 4(2), 145–160.
  38. Maeda, H., Wu, J., Sawa, T., Matsumura, Y., & Hori, K. (2000). Tumor vascular permeability and the EPR effect in macromolecular therapeutics: A review. Journal of Controlled Release, 65(1–2), 271–284. https://doi.org/10.1016/S0168-3659(99)00248-5
  39. Barenholz, Y. (2012). Doxil® — The first FDA-approved nano-drug: Lessons learned. Journal of Controlled Release, 160(2), 117–134. https://doi.org/10.1016/j.jconrel.2012.03.020
  40. Harde, H., Das, M., & Jain, S. (2011). Liposomes: Preparation, characterization and applications. International Journal of Pharmaceutical Sciences Review and Research, 7(2), 1–8.
  41. Bangham, A. D., Standish, M. M., & Watkins, J. C. (1965). Diffusion of univalent ions across the lamellae of swollen phospholipids. Journal of Molecular Biology, 13(1), 238–252. https://doi.org/10.1016/S0022-2836(65)80199-0
  42. Szoka, F., & Papahadjopoulos, D. (1980). Comparative properties and methods of preparation of lipid vesicles (liposomes). Annual Review of Biophysics and Bioengineering, 9, 467–508. https://doi.org/10.1146/annurev.bb.09.060180.002343
  43. Jahn, A., Vreeland, W. N., Gaitan, M., & Locascio, L. E. (2004). Controlled vesicle self-assembly in microfluidic channels with hydrodynamic focusing. Journal of the American Chemical Society, 126(9), 2674–2675. https://doi.org/10.1021/ja039725u
  44. Cegelski, L., & Vreeland, W. N. (2011). Liposome microfluidic technologies for drug delivery. Lab on a Chip, 11(22), 3826–3836. https://doi.org/10.1039/c1lc20543a
  45. New, R. R. C. (1990). Liposomes: A Practical Approach. Oxford University Press.
  46. Mozafari, M. R. (2005). Liposomes: An overview of manufacturing techniques. Cellular and Molecular Biology Letters, 10(4), 711–719.
  47. Immordino, M. L., Dosio, F., & Cattel, L. (2006). Stealth liposomes: Review of the basic science, rationale, and clinical applications, existing and potential. International Journal of Nanomedicine, 1(3), 297–315. https://doi.org/10.2147/nano.2006.1.3.297
  48. Akbarzadeh, A., Rezaei-Sadabady, R., Davaran, S., et al. (2013). Liposome: Classification, preparation, and applications. Nanoscale Research Letters, 8, 102. https://doi.org/10.1186/1556-276X-8-102
  49. Torchilin, V. P. (2005). Recent advances with liposomes as pharmaceutical carriers. Nature Reviews Drug Discovery, 4(2), 145–160. https://doi.org/10.1038/nrd1632
  50. Patil, Y., & Jadhav, S. (2014). Liposomal drug delivery: A comprehensive review. International Journal of Pharmaceutical Research & Development, 6(11), 1–10.
  51. Gupta, A., & Kumar, P. (2010). Liposome drug delivery system: A review. Journal of Advanced Pharmaceutical Technology & Research, 1(4), 374–382. https://doi.org/10.4103/2231-4040.90790
  52. Shaji, J., & Patole, V. (2008). Protein and peptide drug delivery: Oral approaches. Indian Journal of Pharmaceutical Sciences, 70(3), 269–277. https://doi.org/10.4103/0250-474X.43017
  53. Mozafari, M. R., Johnson, C., Hatziantoniou, S., & Demetzos, C. (2008). Nanoliposomes and their applications in food and pharmaceutical sectors. Journal of Liposome Research, 18(3), 309–327. https://doi.org/10.1080/08982100802335964
  54. Deshpande, P., Bisht, S., & Maitra, A. (2009). Evaluation of liposomal anticancer drug formulations. Current Opinion in Investigational Drugs, 10(12), 1334–1341.
  55. Wang, Y., & Yu, H. (2014). Characterization of liposomal drug delivery systems: Particle size and PDI. Journal of Pharmaceutical Analysis, 4(5), 295–302. https://doi.org/10.1016/j.jpha.2014.06.001
  56. Jain, S., & Jain, R. (2015). Zeta potential and stability evaluation of liposomes: A review. International Journal of Pharmaceutical Sciences and Research, 6(3), 882–890. https://doi.org/10.13040/IJPSR.0975-8232.6(3).882-90
  57. Akbarzadeh, A., et al. (2013). Liposome: Classification, preparation, and applications. Nanoscale Research Letters, 8, 102. https://doi.org/10.1186/1556-276X-8-102
  58. ozafari, M. R. (2005). Liposomes: An overview of manufacturing techniques. Cellular and Molecular Biology Letters, 10(4), 711–719.
  59. Kulkarni, A., & Feng, S. S. (2013). In vitro drug release from liposomes: Methods and kinetics. International Journal of Pharmaceutics, 458(1), 116–123. https://doi.org/10.1016/j.ijpharm.2013.09.015
  60. Immordino, M. L., et al. (2006). Stealth liposomes: Review of the basic science, rationale, and clinical applications. International Journal of Nanomedicine, 1(3), 297–315. https://doi.org/10.2147/nano.2006.1.3.297
  61. Deshpande, P., et al. (2009). Evaluation of liposomal anticancer drug formulations. Current Opinion in Investigational Drugs, 10(12), 1334–1341.
  62. Allen, T. M., & Cullis, P. R. (2013). Liposomal drug delivery systems: From concept to clinical applications. Advanced Drug Delivery Reviews, 65(1), 36–48. https://doi.org/10.1016/j.addr.2012.09.037
  63. Bozzuto, G., & Molinari, A. (2015). Liposomes as nanomedical devices. International Journal of Nanomedicine, 10, 975–999. https://doi.org/10.2147/IJN.S68861
  64. Immordino, M. L., Dosio, F., & Cattel, L. (2006). Stealth liposomes: Review of the basic science, rationale, and clinical applications. International Journal of Nanomedicine, 1(3), 297–315. https://doi.org/10.2147/nano.2006.1.3.297
  65. Gabizon, A., Shmeeda, H., & Barenholz, Y. (2003). Pharmacokinetics of pegylated liposomal doxorubicin: Review of animal and human studies. Clinical Pharmacokinetics, 42(5), 419–436. https://doi.org/10.2165/00003088-200342050-00002
  66. Allen, T. M., Hansen, C. B., de Menezes, D., et al. (1991). Liposomes containing synthetic lipid derivatives of poly(ethylene glycol) show prolonged circulation half-lives in vivo. Biochimica et Biophysica Acta (BBA) – Biomembranes, 1066(1), 29–36. https://doi.org/10.1016/0005-2736(91)90143-X
  67. Maeda, H. (2001). The enhanced permeability and retention (EPR) effect in tumor vasculature: The key for tumor-selective drug targeting. Advances in Enzyme Regulation, 41, 189–207. https://doi.org/10.1016/S0065-2571(00)00013-3
  68. Immordino, M. L., et al. (2006). Stealth liposomes: Review of the basic science, rationale, and clinical applications. International Journal of Nanomedicine, 1(3), 297–315. https://doi.org/10.2147/nano.2006.1.3.297
  69. Allen, T. M., & Cullis, P. R. (2013). Liposomal drug delivery systems: From concept to clinical applications. Advanced Drug Delivery Reviews, 65(1), 36–48. https://doi.org/10.1016/j.addr.2012.09.037
  70. Torchilin, V. P. (2005). Recent advances with liposomes as pharmaceutical carriers. Nature Reviews Drug Discovery, 4(2), 145–160. https://doi.org/10.1038/nrd1632
  71. Kummar, S., Chen, A. P., Wright, J., et al. (2007). Phase I study of oral etoposide in patients with advanced solid tumors. Clinical Cancer Research, 13(11), 3377–3383. https://doi.org/10.1158/1078-0432.CCR-06-2708
  72. Hande, K. R. (1998). Etoposide: Four decades of development of a topoisomerase II inhibitor. European Journal of Cancer, 34(10), 1514–1521. https://doi.org/10.1016/S0959-8049(98)00197-2
  73. Gabizon, A., et al. (2003). Pharmacokinetics of pegylated liposomal doxorubicin: Review of animal and human studies. Clinical Pharmacokinetics, 42(5), 419–436. https://doi.org/10.2165/00003088-200342050-00002
  74. Deshpande, P., Bisht, S., & Maitra, A. (2009). Evaluation of liposomal anticancer drug formulations. Current Opinion in Investigational Drugs, 10(12), 1334–1341.
  75. Allen, T. M., Hansen, C. B., de Menezes, D., et al. (1991). Liposomes containing synthetic lipid derivatives of poly(ethylene glycol) show prolonged circulation half-lives in vivo. Biochimica et Biophysica Acta (BBA) – Biomembranes, 1066(1), 29–36. https://doi.org/10.1016/0005-2736(91)90143-X
  76. Mozafari, M. R., Johnson, C., Hatziantoniou, S., & Demetzos, C. (2008). Nanoliposomes and their applications in food and pharmaceutical sectors. Journal of Liposome Research, 18(3), 309–327. https://doi.org/10.1080/08982100802335964
  77. Gelderblom, H., Verweij, J., Nooter, K., & Sparreboom, A. (2001). Cremophor EL: The drawbacks and advantages of vehicle selection for drug formulation. European Journal of Cancer, 37(13), 1590–1598. https://doi.org/10.1016/S0959-8049(01)00142-2
  78. Sparreboom, A., & Verweij, J. (1999). Pharmacokinetics of etoposide and determinants of oral bioavailability. Clinical Pharmacokinetics, 36(6), 447–460. https://doi.org/10.2165/00003088-199936060-00002
  79. McGuire, W. P., Hoskins, W. J., Brady, M. F., et al. (1996). Cyclophosphamide and etoposide in combination therapy. Journal of Clinical Oncology, 14(5), 1646–1655. https://doi.org/10.1200/JCO.1996.14.5.1646
  80. Harde, H., Das, M., & Jain, S. (2011). Liposomes: Preparation, characterization and applications. International Journal of Pharmaceutical Sciences Review and Research, 7(2), 1–8.
  81. Allen, T. M., & Cullis, P. R. (2013). Liposomal drug delivery systems: From concept to clinical applications. Advanced Drug Delivery Reviews, 65(1), 36–48. https://doi.org/10.1016/j.addr.2012.09.037
  82. Bozzuto, G., & Molinari, A. (2015). Liposomes as nanomedical devices. International Journal of Nanomedicine, 10, 975–999. https://doi.org/10.2147/IJN.S68861
  83. Torchilin, V. P. (2005). Recent advances with liposomes as pharmaceutical carriers. Nature Reviews Drug Discovery, 4(2), 145–160. https://doi.org/10.1038/nrd1632
  84. Immordino, M. L., Dosio, F., & Cattel, L. (2006). Stealth liposomes: Review of the basic science, rationale, and clinical applications. International Journal of Nanomedicine, 1(3), 297–315. https://doi.org/10.2147/nano.2006.1.3.297
  85. Kulkarni, A., & Feng, S. S. (2013). In vitro drug release from liposomes: Methods and kinetics. International Journal of Pharmaceutics, 458(1), 116–123. https://doi.org/10.1016/j.ijpharm.2013.09.015
  86. Deshpande, P., et al. (2009). Evaluation of liposomal anticancer drug formulations. Current Opinion in Investigational Drugs, 10(12), 1334–1341.
  87. Gabizon, A., Shmeeda, H., & Barenholz, Y. (2003). Pharmacokinetics of pegylated liposomal doxorubicin: Review of animal and human studies. Clinical Pharmacokinetics, 42(5), 419–436. https://doi.org/10.2165/00003088-200342050-00002
  88. Maeda, H. (2001). The enhanced permeability and retention (EPR) effect in tumor vasculature: The key for tumor-selective drug targeting. Advances in Enzyme Regulation, 41, 189–207. https://doi.org/10.1016/S0065-2571(00)00013-3
  89. Allen, T. M., Hansen, C. B., de Menezes, D., et al. (1991). Liposomes containing synthetic lipid derivatives of poly(ethylene glycol) show prolonged circulation half-lives in vivo. Biochimica et Biophysica Acta (BBA) – Biomembranes, 1066(1), 29–36. https://doi.org/10.1016/0005-2736(91)90143-X
  90. Mozafari, M. R., Johnson, C., Hatziantoniou, S., & Demetzos, C. (2008). Nanoliposomes and their applications in food and pharmaceutical sectors. Journal of Liposome Research, 18(3), 309–327. https://doi.org/10.1080/08982100802335964
  91. Akbarzadeh, A., et al. (2013). Liposome: Classification, preparation, and applications. Nanoscale Research Letters, 8, 102. https://doi.org/10.1186/1556-276X-8-102
  92. Patil, Y., & Jadhav, S. (2014). Liposomal drug delivery: A comprehensive review. International Journal of Pharmaceutical Research & Development, 6(11), 1–10.
  93. Gupta, A., & Kumar, P. (2010). Liposome drug delivery system: A review. Journal of Advanced Pharmaceutical Technology & Research, 1(4), 374–382. https://doi.org/10.4103/2231-4040.90790
  94. Shaji, J., & Patole, V. (2008). Protein and peptide drug delivery: Oral approaches. Indian Journal of Pharmaceutical Sciences, 70(3), 269–277. https://doi.org/10.4103/0250-474X.43017
  95. Harde, H., Das, M., & Jain, S. (2011). Liposomes: Preparation, characterization and applications. International Journal of Pharmaceutical Sciences Review and Research, 7(2), 1–8.
  96. Kulkarni, A., & Feng, S. S. (2013). In vitro drug release from liposomes: Methods and kinetics. International Journal of Pharmaceutics, 458(1), 116–123. https://doi.org/10.1016/j.ijpharm.2013.09.015
  97. Immordino, M. L., et al. (2006). Stealth liposomes: Review of the basic science, rationale, and clinical applications. International Journal of Nanomedicine, 1(3), 297–315. https://doi.org/10.2147/nano.2006.1.3.297
  98. Torchilin, V. P. (2005). Recent advances with liposomes as pharmaceutical carriers. Nature Reviews Drug Discovery, 4(2), 145–160. https://doi.org/10.1038/nrd1632
  99. Allen, T. M., & Cullis, P. R. (2013). Liposomal drug delivery systems: From concept to clinical applications. Advanced Drug Delivery Reviews, 65(1), 36–48. https://doi.org/10.1016/j.addr.2012.09.037
  100. Bozzuto, G., & Molinari, A. (2015). Liposomes as nanomedical devices. International Journal of Nanomedicine, 10, 975–999. https://doi.org/10.2147/IJN.S68861
  101. Gelderblom, H., Verweij, J., Nooter, K., & Sparreboom, A. (2001). Cremophor EL: The drawbacks and advantages of vehicle selection for drug formulation. European Journal of Cancer, 37(13), 1590–1598. https://doi.org/10.1016/S0959-8049(01)00142-2
  102. Sparreboom, A., & Verweij, J. (1999). Pharmacokinetics of etoposide and determinants of oral bioavailability. Clinical Pharmacokinetics, 36(6), 447–460. https://doi.org/10.2165/00003088-199936060-00002
  103. McGuire, W. P., Hoskins, W. J., Brady, M. F., et al. (1996). Cyclophosphamide and etoposide in combination therapy. Journal of Clinical Oncology, 14(5), 1646–1655. https://doi.org/10.1200/JCO.1996.14.5.1646
  104. Allen, T. M., & Cullis, P. R. (2013). Liposomal drug delivery systems: From concept to clinical applications. Advanced Drug Delivery Reviews, 65(1), 36–48. https://doi.org/10.1016/j.addr.2012.09.037
  105. Gabizon, A., Shmeeda, H., & Barenholz, Y. (2003). Pharmacokinetics of pegylated liposomal doxorubicin: Review of animal and human studies. Clinical Pharmacokinetics, 42(5), 419–436. https://doi.org/10.2165/00003088-200342050-00002
  106. Torchilin, V. P. (2005). Recent advances with liposomes as pharmaceutical carriers. Nature Reviews Drug Discovery, 4(2), 145–160. https://doi.org/10.1038/nrd1632
  107. Maeda, H., Wu, J., Sawa, T., Matsumura, Y., & Hori, K. (2000). Tumor vascular permeability and the EPR effect in macromolecular therapeutics: A review. Journal of Controlled Release, 65(1–2), 271–284. https://doi.org/10.1016/S0168-3659(99)00248-5
  108. Holohan, C., Van Schaeybroeck, S., Longley, D. B., & Johnston, P. G. (2013). Cancer drug resistance: An evolving paradigm. Nature Reviews Cancer, 13(10), 714–726. https://doi.org/10.1038/nrc3599
  109. Hande, K. R. (1998). Etoposide: Four decades of development of a topoisomerase II inhibitor. European Journal of Cancer, 34(10), 1514–1521. https://doi.org/10.1016/S0959-8049(98)00197-2
  110. Pommier, Y. (2006). Topoisomerase II inhibitors: Etoposide and beyond. Nature Reviews Cancer, 6(10), 789–802. https://doi.org/10.1038/nrc1977
  111. Kinghorn, A. D., & Pan, L. (2012). Etoposide: Chemistry and pharmacology. Natural Product Reports, 29(8), 967–988. https://doi.org/10.1039/c2np20035f
  112. Kummar, S., Chen, A. P., Wright, J., et al. (2007). Phase I study of oral etoposide in patients with advanced solid tumors. Clinical Cancer Research, 13(11), 3377–3383. https://doi.org/10.1158/1078-0432.CCR-06-2708
  113. Gelderblom, H., Verweij, J., Nooter, K., & Sparreboom, A. (2001). Cremophor EL: The drawbacks and advantages of vehicle selection for drug formulation. European Journal of Cancer, 37(13), 1590–1598. https://doi.org/10.1016/S0959-8049(01)00142-2
  114. Slater, E. P., & Folkman, J. (1984). Podophyllotoxin and derivatives as anticancer agents. Cancer Treatment Reviews, 11(3), 165–182. https://doi.org/10.1016/S0305-7372(84)80005-5
  115. Gewirtz, D. A. (1999). A critical evaluation of the mechanisms of action proposed for the antitumor effects of the anthracycline antibiotics. Biochemical Pharmacology, 57(7), 727–741. https://doi.org/10.1016/S0006-2952(98)00399-1
  116. Barenholz, Y. (2012). Doxil® — The first FDA-approved nano-drug: Lessons learned. Journal of Controlled Release, 160(2), 117–134. https://doi.org/10.1016/j.jconrel.2012.03.020.

Reference

  1. Siegel, R. L., Miller, K. D., & Jemal, A. (2020). Cancer statistics, 2020. CA: A Cancer Journal for Clinicians, 70(1), 7–30. https://doi.org/10.3322/caac.21590
  2. Holohan, C., Van Schaeybroeck, S., Longley, D. B., & Johnston, P. G. (2013). Cancer drug resistance: An evolving paradigm. Nature Reviews Cancer, 13(10), 714–726. https://doi.org/10.1038/nrc3599
  3. Pommier, Y. (2006). Topoisomerase II inhibitors: Etoposide and beyond. Nature Reviews Cancer, 6(10), 789–802. https://doi.org/10.1038/nrc1977
  4. Hande, K. R. (1998). Etoposide: Four decades of development of a topoisomerase II inhibitor. European Journal of Cancer, 34(10), 1514–1521. https://doi.org/10.1016/S0959-8049(98)00197-2
  5. Kummar, S., Chen, A. P., Wright, J., et al. (2007). Phase I study of oral etoposide in patients with advanced solid tumors. Clinical Cancer Research, 13(11), 3377–3383. https://doi.org/10.1158/1078-0432.CCR-06-2708
  6. Gelderblom, H., Verweij, J., Nooter, K., & Sparreboom, A. (2001). Cremophor EL: The drawbacks and advantages of vehicle selection for drug formulation. European Journal of Cancer, 37(13), 1590–1598. https://doi.org/10.1016/S0959-8049(01)00142-2
  7. McGuire, W. P., Hoskins, W. J., Brady, M. F., et al. (1996). Cyclophosphamide and etoposide in combination therapy. Journal of Clinical Oncology, 14(5), 1646–1655. https://doi.org/10.1200/JCO.1996.14.5.1646
  8. Allen, T. M., & Cullis, P. R. (2013). Liposomal drug delivery systems: From concept to clinical applications. Advanced Drug Delivery Reviews, 65(1), 36–48. https://doi.org/10.1016/j.addr.2012.09.037
  9. Bozzuto, G., & Molinari, A. (2015). Liposomes as nanomedical devices. International Journal of Nanomedicine, 10, 975–999. https://doi.org/10.2147/IJN.S68861
  10. Sercombe, L., Veerati, T., Moheimani, F., et al. (2015). Advances and challenges of liposome assisted drug delivery. Frontiers in Pharmacology, 6, 286. https://doi.org/10.3389/fphar.2015.00286
  11. Barenholz, Y. (2012). Doxil® — The first FDA-approved nano-drug: Lessons learned. Journal of Controlled Release, 160(2), 117–134. https://doi.org/10.1016/j.jconrel.2012.03.020
  12. Torchilin, V. P. (2005). Recent advances with liposomes as pharmaceutical carriers. Nature Reviews Drug Discovery, 4(2), 145–160. https://doi.org/10.1038/nrd1632
  13. Maeda, H., Wu, J., Sawa, T., Matsumura, Y., & Hori, K. (2000). Tumor vascular permeability and the EPR effect in macromolecular therapeutics: A review. Journal of Controlled Release, 65(1–2), 271–284. https://doi.org/10.1016/S0168-3659(99)00248-5
  14. Gabizon, A., Shmeeda, H., & Barenholz, Y. (2003). Pharmacokinetics of pegylated liposomal Doxorubicin: Review of animal and human studies. Clinical Pharmacokinetics, 42(5), 419–436. https://doi.org/10.2165/00003088-200342050-00002
  15. Slater, E. P., & Folkman, J. (1984). Podophyllotoxin and derivatives as anticancer agents. Cancer Treatment Reviews, 11(3), 165–182. https://doi.org/10.1016/S0305-7372(84)80005-5
  16. Kinghorn, A. D., & Pan, L. (2012). Etoposide: Chemistry and pharmacology. Natural Product Reports, 29(8), 967–988. https://doi.org/10.1039/c2np20035f
  17. Hande, K. R. (1998). Etoposide: Four decades of development of a topoisomerase II inhibitor. European Journal of Cancer, 34(10), 1514–1521. https://doi.org/10.1016/S0959-8049(98)00197-2
  18. Pommier, Y., Leo, E., Zhang, H., & Marchand, C. (2010). DNA topoisomerases and their poisoning by anticancer and antibacterial drugs. Chemistry & Biology, 17(5), 421–433. https://doi.org/10.1016/j.chembiol.2010.04.012
  19. Gewirtz, D. A. (1999). A critical evaluation of the mechanisms of action proposed for the antitumor effects of the anthracycline antibiotics. Biochemical Pharmacology, 57(7), 727–741. https://doi.org/10.1016/S0006-2952(98)00399-1
  20. Gelderblom, H., Verweij, J., Nooter, K., & Sparreboom, A. (2001). Cremophor EL: The drawbacks and advantages of vehicle selection for drug formulation. European Journal of Cancer, 37(13), 1590–1598. https://doi.org/10.1016/S0959-8049(01)00142-2
  21. Kummar, S., Chen, A. P., Wright, J., et al. (2007). Phase I study of oral etoposide in patients with advanced solid tumors. Clinical Cancer Research, 13(11), 3377–3383. https://doi.org/10.1158/1078-0432.CCR-06-2708
  22. Sparreboom, A., & Verweij, J. (1999). Pharmacokinetics of etoposide and determinants of oral bioavailability. Clinical Pharmacokinetics, 36(6), 447–460. https://doi.org/10.2165/00003088-199936060-00002
  23. Hande, K. R. (1998). Etoposide: Four decades of development of a topoisomerase II inhibitor. European Journal of Cancer, 34(10), 1514–1521. https://doi.org/10.1016/S0959-8049(98)00197-2
  24. Gelderblom, H., et al. (2001). European Journal of Cancer, 37(13), 1590–1598.
  25. McGuire, W. P., et al. (1996). Journal of Clinical Oncology, 14(5), 1646–1655.
  26. Holohan, C., et al. (2013). Nature Reviews Cancer, 13(10), 714–726.
  27. Pommier, Y. (2006). Nature Reviews Cancer, 6(10), 789–802.
  28. Allen, T. M., & Cullis, P. R. (2013). Advanced Drug Delivery Reviews, 65(1), 36–48.
  29. Bozzuto, G., & Molinari, A. (2015). International Journal of Nanomedicine, 10, 975–999.
  30. Bangham, A. D., Standish, M. M., & Watkins, J. C. (1965). Diffusion of univalent ions across the lamellae of swollen phospholipids. Journal of Molecular Biology, 13(1), 238–252. https://doi.org/10.1016/S0022-2836(65)80199-0
  31. Gregoriadis, G. (1995). Engineering liposomes for drug delivery: Progress and problems. Trends in Biotechnology, 13(12), 527–537. https://doi.org/10.1016/S0167-7799(00)88934-4
  32. Allen, T. M. (1994). Liposomal drug formulations: Rationale and prospects for cancer therapy. Drugs, 47(1), 25–36. https://doi.org/10.2165/00003495-199447010-00003
  33. Gabizon, A., et al. (2003). Clinical Pharmacokinetics, 42(5), 419–436.
  34. Lasic, D. D., & Papahadjopoulos, D. (1998). Medical Applications of Liposomes. Elsevier.
  35. Yatvin, M. B., Weinstein, J. N., Dennis, W. H., & Blumenthal, R. (1978). Design of liposomes for enhanced local release of drugs by hyperthermia. Science, 202(4374), 1290–1293. https://doi.org/10.1126/science.688834
  36. Bangham, A. D., et al. (1965). Journal of Molecular Biology, 13(1), 238–252.
  37. Torchilin, V. P. (2005). Nature Reviews Drug Discovery, 4(2), 145–160.
  38. Maeda, H., Wu, J., Sawa, T., Matsumura, Y., & Hori, K. (2000). Tumor vascular permeability and the EPR effect in macromolecular therapeutics: A review. Journal of Controlled Release, 65(1–2), 271–284. https://doi.org/10.1016/S0168-3659(99)00248-5
  39. Barenholz, Y. (2012). Doxil® — The first FDA-approved nano-drug: Lessons learned. Journal of Controlled Release, 160(2), 117–134. https://doi.org/10.1016/j.jconrel.2012.03.020
  40. Harde, H., Das, M., & Jain, S. (2011). Liposomes: Preparation, characterization and applications. International Journal of Pharmaceutical Sciences Review and Research, 7(2), 1–8.
  41. Bangham, A. D., Standish, M. M., & Watkins, J. C. (1965). Diffusion of univalent ions across the lamellae of swollen phospholipids. Journal of Molecular Biology, 13(1), 238–252. https://doi.org/10.1016/S0022-2836(65)80199-0
  42. Szoka, F., & Papahadjopoulos, D. (1980). Comparative properties and methods of preparation of lipid vesicles (liposomes). Annual Review of Biophysics and Bioengineering, 9, 467–508. https://doi.org/10.1146/annurev.bb.09.060180.002343
  43. Jahn, A., Vreeland, W. N., Gaitan, M., & Locascio, L. E. (2004). Controlled vesicle self-assembly in microfluidic channels with hydrodynamic focusing. Journal of the American Chemical Society, 126(9), 2674–2675. https://doi.org/10.1021/ja039725u
  44. Cegelski, L., & Vreeland, W. N. (2011). Liposome microfluidic technologies for drug delivery. Lab on a Chip, 11(22), 3826–3836. https://doi.org/10.1039/c1lc20543a
  45. New, R. R. C. (1990). Liposomes: A Practical Approach. Oxford University Press.
  46. Mozafari, M. R. (2005). Liposomes: An overview of manufacturing techniques. Cellular and Molecular Biology Letters, 10(4), 711–719.
  47. Immordino, M. L., Dosio, F., & Cattel, L. (2006). Stealth liposomes: Review of the basic science, rationale, and clinical applications, existing and potential. International Journal of Nanomedicine, 1(3), 297–315. https://doi.org/10.2147/nano.2006.1.3.297
  48. Akbarzadeh, A., Rezaei-Sadabady, R., Davaran, S., et al. (2013). Liposome: Classification, preparation, and applications. Nanoscale Research Letters, 8, 102. https://doi.org/10.1186/1556-276X-8-102
  49. Torchilin, V. P. (2005). Recent advances with liposomes as pharmaceutical carriers. Nature Reviews Drug Discovery, 4(2), 145–160. https://doi.org/10.1038/nrd1632
  50. Patil, Y., & Jadhav, S. (2014). Liposomal drug delivery: A comprehensive review. International Journal of Pharmaceutical Research & Development, 6(11), 1–10.
  51. Gupta, A., & Kumar, P. (2010). Liposome drug delivery system: A review. Journal of Advanced Pharmaceutical Technology & Research, 1(4), 374–382. https://doi.org/10.4103/2231-4040.90790
  52. Shaji, J., & Patole, V. (2008). Protein and peptide drug delivery: Oral approaches. Indian Journal of Pharmaceutical Sciences, 70(3), 269–277. https://doi.org/10.4103/0250-474X.43017
  53. Mozafari, M. R., Johnson, C., Hatziantoniou, S., & Demetzos, C. (2008). Nanoliposomes and their applications in food and pharmaceutical sectors. Journal of Liposome Research, 18(3), 309–327. https://doi.org/10.1080/08982100802335964
  54. Deshpande, P., Bisht, S., & Maitra, A. (2009). Evaluation of liposomal anticancer drug formulations. Current Opinion in Investigational Drugs, 10(12), 1334–1341.
  55. Wang, Y., & Yu, H. (2014). Characterization of liposomal drug delivery systems: Particle size and PDI. Journal of Pharmaceutical Analysis, 4(5), 295–302. https://doi.org/10.1016/j.jpha.2014.06.001
  56. Jain, S., & Jain, R. (2015). Zeta potential and stability evaluation of liposomes: A review. International Journal of Pharmaceutical Sciences and Research, 6(3), 882–890. https://doi.org/10.13040/IJPSR.0975-8232.6(3).882-90
  57. Akbarzadeh, A., et al. (2013). Liposome: Classification, preparation, and applications. Nanoscale Research Letters, 8, 102. https://doi.org/10.1186/1556-276X-8-102
  58. ozafari, M. R. (2005). Liposomes: An overview of manufacturing techniques. Cellular and Molecular Biology Letters, 10(4), 711–719.
  59. Kulkarni, A., & Feng, S. S. (2013). In vitro drug release from liposomes: Methods and kinetics. International Journal of Pharmaceutics, 458(1), 116–123. https://doi.org/10.1016/j.ijpharm.2013.09.015
  60. Immordino, M. L., et al. (2006). Stealth liposomes: Review of the basic science, rationale, and clinical applications. International Journal of Nanomedicine, 1(3), 297–315. https://doi.org/10.2147/nano.2006.1.3.297
  61. Deshpande, P., et al. (2009). Evaluation of liposomal anticancer drug formulations. Current Opinion in Investigational Drugs, 10(12), 1334–1341.
  62. Allen, T. M., & Cullis, P. R. (2013). Liposomal drug delivery systems: From concept to clinical applications. Advanced Drug Delivery Reviews, 65(1), 36–48. https://doi.org/10.1016/j.addr.2012.09.037
  63. Bozzuto, G., & Molinari, A. (2015). Liposomes as nanomedical devices. International Journal of Nanomedicine, 10, 975–999. https://doi.org/10.2147/IJN.S68861
  64. Immordino, M. L., Dosio, F., & Cattel, L. (2006). Stealth liposomes: Review of the basic science, rationale, and clinical applications. International Journal of Nanomedicine, 1(3), 297–315. https://doi.org/10.2147/nano.2006.1.3.297
  65. Gabizon, A., Shmeeda, H., & Barenholz, Y. (2003). Pharmacokinetics of pegylated liposomal doxorubicin: Review of animal and human studies. Clinical Pharmacokinetics, 42(5), 419–436. https://doi.org/10.2165/00003088-200342050-00002
  66. Allen, T. M., Hansen, C. B., de Menezes, D., et al. (1991). Liposomes containing synthetic lipid derivatives of poly(ethylene glycol) show prolonged circulation half-lives in vivo. Biochimica et Biophysica Acta (BBA) – Biomembranes, 1066(1), 29–36. https://doi.org/10.1016/0005-2736(91)90143-X
  67. Maeda, H. (2001). The enhanced permeability and retention (EPR) effect in tumor vasculature: The key for tumor-selective drug targeting. Advances in Enzyme Regulation, 41, 189–207. https://doi.org/10.1016/S0065-2571(00)00013-3
  68. Immordino, M. L., et al. (2006). Stealth liposomes: Review of the basic science, rationale, and clinical applications. International Journal of Nanomedicine, 1(3), 297–315. https://doi.org/10.2147/nano.2006.1.3.297
  69. Allen, T. M., & Cullis, P. R. (2013). Liposomal drug delivery systems: From concept to clinical applications. Advanced Drug Delivery Reviews, 65(1), 36–48. https://doi.org/10.1016/j.addr.2012.09.037
  70. Torchilin, V. P. (2005). Recent advances with liposomes as pharmaceutical carriers. Nature Reviews Drug Discovery, 4(2), 145–160. https://doi.org/10.1038/nrd1632
  71. Kummar, S., Chen, A. P., Wright, J., et al. (2007). Phase I study of oral etoposide in patients with advanced solid tumors. Clinical Cancer Research, 13(11), 3377–3383. https://doi.org/10.1158/1078-0432.CCR-06-2708
  72. Hande, K. R. (1998). Etoposide: Four decades of development of a topoisomerase II inhibitor. European Journal of Cancer, 34(10), 1514–1521. https://doi.org/10.1016/S0959-8049(98)00197-2
  73. Gabizon, A., et al. (2003). Pharmacokinetics of pegylated liposomal doxorubicin: Review of animal and human studies. Clinical Pharmacokinetics, 42(5), 419–436. https://doi.org/10.2165/00003088-200342050-00002
  74. Deshpande, P., Bisht, S., & Maitra, A. (2009). Evaluation of liposomal anticancer drug formulations. Current Opinion in Investigational Drugs, 10(12), 1334–1341.
  75. Allen, T. M., Hansen, C. B., de Menezes, D., et al. (1991). Liposomes containing synthetic lipid derivatives of poly(ethylene glycol) show prolonged circulation half-lives in vivo. Biochimica et Biophysica Acta (BBA) – Biomembranes, 1066(1), 29–36. https://doi.org/10.1016/0005-2736(91)90143-X
  76. Mozafari, M. R., Johnson, C., Hatziantoniou, S., & Demetzos, C. (2008). Nanoliposomes and their applications in food and pharmaceutical sectors. Journal of Liposome Research, 18(3), 309–327. https://doi.org/10.1080/08982100802335964
  77. Gelderblom, H., Verweij, J., Nooter, K., & Sparreboom, A. (2001). Cremophor EL: The drawbacks and advantages of vehicle selection for drug formulation. European Journal of Cancer, 37(13), 1590–1598. https://doi.org/10.1016/S0959-8049(01)00142-2
  78. Sparreboom, A., & Verweij, J. (1999). Pharmacokinetics of etoposide and determinants of oral bioavailability. Clinical Pharmacokinetics, 36(6), 447–460. https://doi.org/10.2165/00003088-199936060-00002
  79. McGuire, W. P., Hoskins, W. J., Brady, M. F., et al. (1996). Cyclophosphamide and etoposide in combination therapy. Journal of Clinical Oncology, 14(5), 1646–1655. https://doi.org/10.1200/JCO.1996.14.5.1646
  80. Harde, H., Das, M., & Jain, S. (2011). Liposomes: Preparation, characterization and applications. International Journal of Pharmaceutical Sciences Review and Research, 7(2), 1–8.
  81. Allen, T. M., & Cullis, P. R. (2013). Liposomal drug delivery systems: From concept to clinical applications. Advanced Drug Delivery Reviews, 65(1), 36–48. https://doi.org/10.1016/j.addr.2012.09.037
  82. Bozzuto, G., & Molinari, A. (2015). Liposomes as nanomedical devices. International Journal of Nanomedicine, 10, 975–999. https://doi.org/10.2147/IJN.S68861
  83. Torchilin, V. P. (2005). Recent advances with liposomes as pharmaceutical carriers. Nature Reviews Drug Discovery, 4(2), 145–160. https://doi.org/10.1038/nrd1632
  84. Immordino, M. L., Dosio, F., & Cattel, L. (2006). Stealth liposomes: Review of the basic science, rationale, and clinical applications. International Journal of Nanomedicine, 1(3), 297–315. https://doi.org/10.2147/nano.2006.1.3.297
  85. Kulkarni, A., & Feng, S. S. (2013). In vitro drug release from liposomes: Methods and kinetics. International Journal of Pharmaceutics, 458(1), 116–123. https://doi.org/10.1016/j.ijpharm.2013.09.015
  86. Deshpande, P., et al. (2009). Evaluation of liposomal anticancer drug formulations. Current Opinion in Investigational Drugs, 10(12), 1334–1341.
  87. Gabizon, A., Shmeeda, H., & Barenholz, Y. (2003). Pharmacokinetics of pegylated liposomal doxorubicin: Review of animal and human studies. Clinical Pharmacokinetics, 42(5), 419–436. https://doi.org/10.2165/00003088-200342050-00002
  88. Maeda, H. (2001). The enhanced permeability and retention (EPR) effect in tumor vasculature: The key for tumor-selective drug targeting. Advances in Enzyme Regulation, 41, 189–207. https://doi.org/10.1016/S0065-2571(00)00013-3
  89. Allen, T. M., Hansen, C. B., de Menezes, D., et al. (1991). Liposomes containing synthetic lipid derivatives of poly(ethylene glycol) show prolonged circulation half-lives in vivo. Biochimica et Biophysica Acta (BBA) – Biomembranes, 1066(1), 29–36. https://doi.org/10.1016/0005-2736(91)90143-X
  90. Mozafari, M. R., Johnson, C., Hatziantoniou, S., & Demetzos, C. (2008). Nanoliposomes and their applications in food and pharmaceutical sectors. Journal of Liposome Research, 18(3), 309–327. https://doi.org/10.1080/08982100802335964
  91. Akbarzadeh, A., et al. (2013). Liposome: Classification, preparation, and applications. Nanoscale Research Letters, 8, 102. https://doi.org/10.1186/1556-276X-8-102
  92. Patil, Y., & Jadhav, S. (2014). Liposomal drug delivery: A comprehensive review. International Journal of Pharmaceutical Research & Development, 6(11), 1–10.
  93. Gupta, A., & Kumar, P. (2010). Liposome drug delivery system: A review. Journal of Advanced Pharmaceutical Technology & Research, 1(4), 374–382. https://doi.org/10.4103/2231-4040.90790
  94. Shaji, J., & Patole, V. (2008). Protein and peptide drug delivery: Oral approaches. Indian Journal of Pharmaceutical Sciences, 70(3), 269–277. https://doi.org/10.4103/0250-474X.43017
  95. Harde, H., Das, M., & Jain, S. (2011). Liposomes: Preparation, characterization and applications. International Journal of Pharmaceutical Sciences Review and Research, 7(2), 1–8.
  96. Kulkarni, A., & Feng, S. S. (2013). In vitro drug release from liposomes: Methods and kinetics. International Journal of Pharmaceutics, 458(1), 116–123. https://doi.org/10.1016/j.ijpharm.2013.09.015
  97. Immordino, M. L., et al. (2006). Stealth liposomes: Review of the basic science, rationale, and clinical applications. International Journal of Nanomedicine, 1(3), 297–315. https://doi.org/10.2147/nano.2006.1.3.297
  98. Torchilin, V. P. (2005). Recent advances with liposomes as pharmaceutical carriers. Nature Reviews Drug Discovery, 4(2), 145–160. https://doi.org/10.1038/nrd1632
  99. Allen, T. M., & Cullis, P. R. (2013). Liposomal drug delivery systems: From concept to clinical applications. Advanced Drug Delivery Reviews, 65(1), 36–48. https://doi.org/10.1016/j.addr.2012.09.037
  100. Bozzuto, G., & Molinari, A. (2015). Liposomes as nanomedical devices. International Journal of Nanomedicine, 10, 975–999. https://doi.org/10.2147/IJN.S68861
  101. Gelderblom, H., Verweij, J., Nooter, K., & Sparreboom, A. (2001). Cremophor EL: The drawbacks and advantages of vehicle selection for drug formulation. European Journal of Cancer, 37(13), 1590–1598. https://doi.org/10.1016/S0959-8049(01)00142-2
  102. Sparreboom, A., & Verweij, J. (1999). Pharmacokinetics of etoposide and determinants of oral bioavailability. Clinical Pharmacokinetics, 36(6), 447–460. https://doi.org/10.2165/00003088-199936060-00002
  103. McGuire, W. P., Hoskins, W. J., Brady, M. F., et al. (1996). Cyclophosphamide and etoposide in combination therapy. Journal of Clinical Oncology, 14(5), 1646–1655. https://doi.org/10.1200/JCO.1996.14.5.1646
  104. Allen, T. M., & Cullis, P. R. (2013). Liposomal drug delivery systems: From concept to clinical applications. Advanced Drug Delivery Reviews, 65(1), 36–48. https://doi.org/10.1016/j.addr.2012.09.037
  105. Gabizon, A., Shmeeda, H., & Barenholz, Y. (2003). Pharmacokinetics of pegylated liposomal doxorubicin: Review of animal and human studies. Clinical Pharmacokinetics, 42(5), 419–436. https://doi.org/10.2165/00003088-200342050-00002
  106. Torchilin, V. P. (2005). Recent advances with liposomes as pharmaceutical carriers. Nature Reviews Drug Discovery, 4(2), 145–160. https://doi.org/10.1038/nrd1632
  107. Maeda, H., Wu, J., Sawa, T., Matsumura, Y., & Hori, K. (2000). Tumor vascular permeability and the EPR effect in macromolecular therapeutics: A review. Journal of Controlled Release, 65(1–2), 271–284. https://doi.org/10.1016/S0168-3659(99)00248-5
  108. Holohan, C., Van Schaeybroeck, S., Longley, D. B., & Johnston, P. G. (2013). Cancer drug resistance: An evolving paradigm. Nature Reviews Cancer, 13(10), 714–726. https://doi.org/10.1038/nrc3599
  109. Hande, K. R. (1998). Etoposide: Four decades of development of a topoisomerase II inhibitor. European Journal of Cancer, 34(10), 1514–1521. https://doi.org/10.1016/S0959-8049(98)00197-2
  110. Pommier, Y. (2006). Topoisomerase II inhibitors: Etoposide and beyond. Nature Reviews Cancer, 6(10), 789–802. https://doi.org/10.1038/nrc1977
  111. Kinghorn, A. D., & Pan, L. (2012). Etoposide: Chemistry and pharmacology. Natural Product Reports, 29(8), 967–988. https://doi.org/10.1039/c2np20035f
  112. Kummar, S., Chen, A. P., Wright, J., et al. (2007). Phase I study of oral etoposide in patients with advanced solid tumors. Clinical Cancer Research, 13(11), 3377–3383. https://doi.org/10.1158/1078-0432.CCR-06-2708
  113. Gelderblom, H., Verweij, J., Nooter, K., & Sparreboom, A. (2001). Cremophor EL: The drawbacks and advantages of vehicle selection for drug formulation. European Journal of Cancer, 37(13), 1590–1598. https://doi.org/10.1016/S0959-8049(01)00142-2
  114. Slater, E. P., & Folkman, J. (1984). Podophyllotoxin and derivatives as anticancer agents. Cancer Treatment Reviews, 11(3), 165–182. https://doi.org/10.1016/S0305-7372(84)80005-5
  115. Gewirtz, D. A. (1999). A critical evaluation of the mechanisms of action proposed for the antitumor effects of the anthracycline antibiotics. Biochemical Pharmacology, 57(7), 727–741. https://doi.org/10.1016/S0006-2952(98)00399-1
  116. Barenholz, Y. (2012). Doxil® — The first FDA-approved nano-drug: Lessons learned. Journal of Controlled Release, 160(2), 117–134. https://doi.org/10.1016/j.jconrel.2012.03.020.

Photo
Rushikesh Nirgude
Corresponding author

Matoshri College of Pharmacy, Eklahare, Nashik

Photo
Vedika Pagar
Co-author

Matoshri College of Pharmacy, Eklahare, Nashik

Photo
Ragini Kadvekar
Co-author

Matoshri College of Pharmacy, Eklahare, Nashik

Rushikesh Nirgude*, Vedika Pagar, Ragini Kadvekar, Formulation and Evaluation of Etoposide Liposomal Drug Delivery Systems, Int. J. Sci. R. Tech., 2025, 2 (10), 410-427. https://doi.org/10.5281/zenodo.17444421

Related Articles
From Localization to Connectomics: A Contemporary View of Human Brain Structure ...
Arnab Roy, Mahesh Kumar Yadav, Anchal Kumari , Karishma Kumari , Amit Kumar Prajapati , Vivek Prajap...
Pharmacy Supply Chain Dynamics and the Importance of Pharmacists in Retail Medic...
Arnab Roy, Mahesh Kumar Yadav , Md. Asif , Lawkush Kumar Prajapati , Md. Kaif , Makhmur Hayat , Rup...
More related articles
Pharmacy Supply Chain Dynamics and the Importance of Pharmacists in Retail Medic...
Arnab Roy, Mahesh Kumar Yadav , Md. Asif , Lawkush Kumar Prajapati , Md. Kaif , Makhmur Hayat , Rup...
Transforming Pharmacy Automation: The Role of Robotics and Artificial Intelligen...
Dipak Bhingardeve, Yuvraj Amale, Atul Kadam, Amit Atugade, Sanket Patil, ...
Pharmacy Supply Chain Dynamics and the Importance of Pharmacists in Retail Medic...
Arnab Roy, Mahesh Kumar Yadav , Md. Asif , Lawkush Kumar Prajapati , Md. Kaif , Makhmur Hayat , Rup...
Transforming Pharmacy Automation: The Role of Robotics and Artificial Intelligen...
Dipak Bhingardeve, Yuvraj Amale, Atul Kadam, Amit Atugade, Sanket Patil, ...