1,2,3 SSP Shikshan Sanstha’s Siddhi College of Pharmacy, Chikhali, Pune, Maharashtra, India 411062
4 SSSPM Shree Babasaheb Gharfalkar College of Pharmacy, Nachangaon, Pulgaon , Maharashtra, India 442302
Ovarian cancer (OC) remains the deadliest of the gynecologic malignancies, largely because more than 70?% of patients present with advanced?stage disease and quickly acquire resistance to platinum?based chemotherapy. Liposomes spherical vesicles composed of one or more phospholipid bilayers were the first nanocarriers to translate from bench to bedside. By encapsulating cytotoxics or nucleic acids, liposomes can enhance pharmacokinetics, improve tumor deposition, and mitigate systemic toxicities. Pegylated liposomal doxorubicin (PLD) is now a backbone therapy for recurrent OC, and a new generation of trigger?responsive, ligand?targeted, and gene?loaded liposomes are poised to address persistent challenges such as intraperitoneal dissemination and genomic heterogeneity. This review summarizes the physicochemical foundations of liposome design, surveys current clinical and preclinical liposomal formulations in OC, highlights emerging strategies (stimuli?responsive, multi?drug, and immuno?activating systems), and discusses translational hurdles and future directions. Collectively, the field is moving toward precision nanomedicine in which liposomes function not only as drug ferries but also as programmable mini organs capable of sensing and modulating the tumor microenvironment.
Ovarian cancer accounts for approximately 313,?000 new diagnoses and 207,?000 deaths worldwide annually, making it the eighth leading cause of female cancer mortality [1]. Standard front?line management surgical cytoreduction plus platinum/taxane chemotherapy achieves high initial response rates, yet more than 80?% of patients relapse within two years [2]. Immunotherapies and molecularly targeted agents (e.g., PARP inhibitors) are now integral, but cumulative toxicities, dose?limiting myelosuppression, and poor intraperitoneal drug penetration still curtail durable control. Nanocarrier?mediated delivery, spearheaded by liposomes, emerged in the 1990s to re?engineer drug exposure profiles and exploit the enhanced permeability and retention (EPR) effect of tumors [3].
Liposomes consist of an aqueous core surrounded by lipid bilayer(s) that can incorporate hydrophilic and hydrophobic payloads. Stealth coatings (typically 2?kDa polyethylene glycol) extend circulation half?life from minutes to roughly 30?hours, while surface ligands such as folate or anti?EphA2 promote active targeting of OC cells [4]. To date, more than 15 liposomal drugs have received regulatory approval across indications, two of which PLD (Doxil/Caelyx) and liposomal irinotecan (Onivyde) have demonstrated survival benefit in solid tumors. OC, with its extensive peritoneal vascular fenestrations and high folate?receptor expression, is ideally suited for liposomal interventions [5].
This review first revisits liposome engineering principles before turning to OC biology. We then integrate clinical evidence for marketed and investigational liposomal agents, examine preclinical advances in gene and immune modulation, and critique translational challenges such as EPR heterogeneity and immune clearance. Finally, we outline future research priorities, including combinatorial nanomedicine and image?guided intraperitoneal delivery.
2. Fundamentals of Liposome Technology:
2.1. Composition and Architecture:
Figure 1. Liposome structure showing the hydrophilic core and lipid bilayer.
A canonical liposome is built from phosphatidylcholine and cholesterol in a 55:45 mol% ratio, affording a liquid ordered bilayer with low permeability to serum proteins. Manipulating lipid composition modulates rigidity, fusogenicity, and release kinetics [6]. High transition temperature lipids such as DSPC allow lysolipid thermosensitive liposomes that burst at 40-42?°C, enabling on-demand release during focused hyperthermia. Cationic lipids (e.g., DOTAP) electrostatically bind nucleic acids but provoke complement activation; ionizable lipids circumvent this by remaining neutral at physiologic pH and protonating in the acidic endosome [7].
2.2. Advantages of Liposomes:
2.3. Disadvantages of Liposomes:
2.4. Manufacturing Methods:
Thin?film hydration with extrusion remains ubiquitous for lab?scale batches, yet pharmaceutical?grade products rely on ethanol?injection or microfluidic mixing to ensure narrow polydispersity (<?0.1) and scalable throughput. Remote loading using a transmembrane (NH?)?SO? or pH gradient routinely achieves greater than 95?% doxorubicin entrapment at drug?to?lipid ratios of 0.2 (w/w) [8].
Manufacturing methods of liposomes vary based on the desired size, lamellarity (number of bilayers), drug encapsulation efficiency, and scalability. Below is a comprehensive overview of the commonly used liposome manufacturing techniques:
I. Thin Film Hydration (Bangham Method)
II. Reverse Phase Evaporation Method (REV)
III. Ethanol Injection Method
IV. Microfluidics Method
V. Freeze–Thaw Method
VI. Detergent Removal Method
Table No. 1 Comparison Table of Liposome Manufacturing Methods
Sr.No. |
Method |
Vesicle Type |
Encapsulation Efficiency |
Scalability |
Complexity |
1. |
Thin Film Hydration |
MLV, SUV |
Low (hydrophilic drugs) |
Low |
Low |
2. |
Reverse Phase Evaporation |
LUV |
High |
Moderate |
Moderate |
3. |
Ethanol Injection |
SUV |
Low |
Moderate |
Low |
4. |
Microfluidics |
SUV, LUV |
Moderate–High |
High |
High |
5. |
Freeze–Thaw |
MLV → LUV |
Improved |
Low |
Moderate |
6. |
Detergent Removal |
SUV |
Moderate |
Low–Moderate |
High |
2.5. Pharmacokinetics and Biodistribution:
Stealth liposomes evade opsonization and reduce hepatic capture, prolonging area under the curve and exploiting the leaky neovasculature of tumors (fenestrae 200–800?nm). However, EPR is heterogenous across patients and even within a single tumor [9]. Strategies to boost deposition include low?dose TNF?α, ultrasound?mediated microbubble disruption, and ligand?mediated endocytosis.
Liposomes, as nanoscale drug carriers, significantly alter the pharmacokinetics (PK) and biodistribution (BD) profiles of encapsulated therapeutics compared to their free forms. Their size, composition, surface characteristics, and route of administration influence their in vivo behavior, enhancing therapeutic efficacy and minimizing systemic toxicity.
2.5.1. Pharmacokinetics of Liposomes
Liposomes modify all four pillars of pharmacokinetics - absorption, distribution, metabolism, and excretion (ADME) to improve drug performance.
2.5.2. Factors Influencing Liposome Pharmacokinetics
Table No. 2 Factors Influencing Liposome Pharmacokinetics[53]
Factor |
Effect on PK |
Size |
Larger liposomes (>200 nm) are cleared more rapidly. |
Charge |
Neutral or slightly negative charges favor longevity. |
Surface Modification |
PEGylation prolongs circulation and reduces RES uptake. |
Lipid Composition |
Saturated lipids and cholesterol enhance stability. |
2.5.3. Biodistribution of Liposomes
Tumors, due to defective vasculature and impaired lymphatic drainage, allow nanoscale liposomes to accumulate preferentially—a phenomenon called the EPR effect [51].
Surface modification with ligands such as antibodies, peptides, aptamers, or sugars enables receptor-mediated endocytosis, increasing specificity and uptake by target cells [54].
Unmodified liposomes are rapidly sequestered by macrophages in the liver, spleen, and lungs. PEGylation reduces this uptake, but some accumulation in these organs still occurs [55].
2.5.4. Organ Distribution Overview
Table No. 3 Organ Distribution Overview
Organ |
Liposome Accumulation (↑ High / ↓ Low) |
Liver |
↑ (especially Kupffer cell uptake) |
Spleen |
↑ |
Lung |
↑ (in some formulations) |
Tumor tissue |
↑ (via EPR and active targeting) |
Kidney |
↓ (limited renal clearance) |
Heart/Brain |
↓ (blood-brain barrier limits entry) |
2.5.5. Intracellular Fate
After reaching the target tissue, liposomes may be:
2.5.6. Clinical Relevance
Liposomal drugs such as Doxil® (pegylated liposomal doxorubicin) and Ambisome® (liposomal amphotericin B) demonstrate:
Their pharmacokinetic advantages result in higher area under the curve (AUC), lower peak plasma concentrations (Cmax), and prolonged half-life, especially compared to conventional free drugs [6].
2.6. Safety Profile:
PEGylation dramatically lowers infusion?related reactions but introduces accelerated blood clearance upon repeat dosing. Hand?foot syndrome and mucositis, while milder than the neutropenia and alopecia typical of free doxorubicin, remain dose?limiting for PLD. Novel hydrophilic coatings and intermittent dosing schedules mitigate hand?foot syndrome incidence to below 10?% [10].
Liposomes are among the most extensively studied nanocarriers for drug delivery, particularly in oncology, due to their biocompatibility, reduced systemic toxicity, and ability to encapsulate both hydrophilic and hydrophobic drugs. However, like any therapeutic system, they are not devoid of adverse effects and safety considerations.
2.6.1. Biocompatibility and Immunogenicity
Liposomes, particularly those composed of natural or synthetic phospholipids, exhibit excellent biocompatibility. Their similarity to biological membranes contributes to low immunogenicity. However, certain formulations can still provoke immune responses:
2.6.2. Toxicity
2.6.3. Influence of Physicochemical Properties on Safety
2.6.4. Safety in Special Populations
2.6.5. Clinical Evidence
2.6.6. Regulatory and Long-Term Safety
3. Ovarian Cancer Biology and Treatment Landscape:
Figure 2. Ovarian Cancer Biology
Epithelial ovarian cancer (EOC) constitutes approximately 90?% of cases and is subdivided into high?grade serous, endometrioid, clear?cell, mucinous, and low?grade serous subtypes [2]. TP53 mutations dominate high?grade serous carcinogenesis, accompanied by homologous?recombination deficiency in about 50?% of tumors, rendering them sensitive to PARP inhibition. Peritoneal dissemination, ascites formation, and immune?suppressive macrophages characterize advanced disease.
First?line therapy comprises optimal debulking surgery plus carboplatin and paclitaxel, sometimes augmented by bevacizumab. Despite an 80–90?% objective response rate, median progression?free survival is roughly 18 months. Platinum?resistant relapse carries a median overall survival of 12 months, prompting exploration of non?cross?resistant agents such as PLD. The peritoneal?plasma barrier and dose?limiting myelosuppression often halt dose intensification; hence nanocarrier delivery to intensify intratumoral exposures while sparing marrow is compelling.
Unique features of OC that favor liposomal strategies include folate receptor?α overexpression on most serous tumors, large peritoneal surface area that promotes nanoparticle settling, and tumor ascites acting as a drug reservoir. In summary, OC pathobiology both necessitates and facilitates liposome?mediated interventions.
4. Liposomes Used in Ovarian Cancer:
Figure 3. Timeline depicting key liposomal formulations developed for ovarian cancer treatment.
Liposomes have revolutionized the field of drug delivery, offering promising therapeutic avenues particularly in oncology. In ovarian cancer (OC), where the disease is often diagnosed at an advanced stage with extensive peritoneal spread, liposomal technologies have found unique utility. The need for enhanced drug delivery platforms stems from challenges such as systemic toxicity of conventional chemotherapeutics, limited intraperitoneal drug penetration, chemoresistance, and immunosuppressive tumor microenvironments. Liposomes provide a means of delivering higher concentrations of therapeutic agents directly to tumor tissues, minimizing off-target effects and improving the therapeutic index.
The most well-established liposomal formulation in OC therapy is Pegylated Liposomal Doxorubicin (PLD), known by the brand names Doxil® and Caelyx®. The encapsulation of doxorubicin within PEGylated liposomes enhances its plasma half-life and improves its accumulation at tumor sites via the enhanced permeability and retention (EPR) effect. Numerous clinical trials have demonstrated the efficacy of PLD in recurrent and platinum-resistant ovarian cancer, showing an objective response rate of 15–20% and a favorable toxicity profile characterized by reduced cardiotoxicity and alopecia [1,2]. PLD is often used in combination with carboplatin, with meta-analyses supporting its superiority over traditional paclitaxel–carboplatin regimens in prolonging progression-free survival [3].
Lipoplatin®, a liposomal formulation of cisplatin, addresses the critical limitation of nephrotoxicity associated with free cisplatin. Its lipid encapsulation reduces renal accumulation, enabling safer administration at effective doses. Phase II studies have demonstrated non-inferiority in clinical outcomes with a better renal safety profile [4]. Lipoplatin has shown synergy with paclitaxel and is being evaluated for use in platinum-sensitive and platinum-resistant cases alike.
LEP-ETU, a liposomal formulation of paclitaxel, is under development to reduce hypersensitivity reactions and eliminate the need for Cremophor EL, a solvent responsible for severe infusion reactions. LEP-ETU exhibits greater stability and sustained release kinetics, with enhanced bioavailability in the peritoneal cavity an ideal feature for OC management [5].
Folate-receptor-targeted liposomes exploit the overexpression of folate receptor-α (FR-α) in over 80% of epithelial OC cases. By conjugating folate to the liposome surface, drug payloads such as doxorubicin, paclitaxel, and PARP inhibitors are directed to tumor cells with high specificity. Preclinical studies have shown enhanced internalization and cytotoxicity, while clinical trials are assessing efficacy and safety [6,7]. Targeted liposomes improve drug accumulation in tumors, reduce systemic distribution, and demonstrate lower off-target effects.
Another advancement includes the use of immuno-liposomes, liposomes engineered to deliver immunotherapies such as anti-PD-1, anti-CTLA-4, and immune-activating cytokines like IL-2 or GM-CSF. These liposomes aim to modulate the tumor microenvironment, enhancing antigen presentation and overcoming immune evasion. In OC, where immunotherapy responses have historically been limited, this approach shows promise in reactivating anti-tumor immunity [8,9].
Gene therapy liposomes represent a novel strategy aimed at modulating genetic pathways involved in chemoresistance and tumor progression. Cationic liposomes have been employed to deliver tumor suppressor genes (e.g., p53, BRCA1/2), siRNA against drug resistance genes (e.g., MDR1), and CRISPR-Cas9 elements targeting oncogenic mutations. Several animal studies have demonstrated tumor regression, delayed recurrence, and increased sensitivity to platinum-based chemotherapy following gene delivery via liposomes [10,11]. Ionizable lipid nanoparticles provide superior transfection efficiency and reduced systemic toxicity compared to traditional cationic systems.
Furthermore, exosome-mimetic liposomes are an emerging class of hybrid nanocarriers that incorporate exosomal membrane proteins to enhance biocompatibility and tumor tropism. These vesicles mimic the natural targeting abilities of exosomes while leveraging the stability and drug-loading capacity of liposomes. Studies in OC models indicate increased peritoneal retention and effective delivery of paclitaxel and siRNA payloads to drug-resistant tumor cells [12].
Stimuli-responsive liposomes, designed to release their contents in response to environmental cues such as low pH, enzymatic activity, or elevated glutathione levels, are also under investigation. Given the acidic tumor microenvironment and the presence of reductive agents in the intracellular space, these liposomes provide controlled and site-specific release of therapeutic agents. Preclinical trials have confirmed their capacity to reduce systemic exposure and enhance tumor-specific cytotoxicity [13].
Moreover, multimodal liposomes incorporating imaging agents such as gadolinium for MRI or copper-64 for PET have been developed to enable real-time tracking of liposome biodistribution and therapeutic monitoring. Such theranostic systems could facilitate personalized dosing and improve response assessment in OC patients [14].
Sonodynamic and photodynamic liposomes (e.g., porphysomes) are another exciting innovation, offering the ability to generate cytotoxic reactive oxygen species upon ultrasound or light exposure. These platforms allow for externally controlled, spatially confined therapy and have shown promise in intraperitoneal OC models, where precision is key to avoid damaging healthy tissue [15].
Taken together, the liposomal systems used in ovarian cancer ranging from classic chemotherapeutic carriers to sophisticated gene and immune modulators represent a diverse and rapidly evolving toolkit. As understanding of tumor biology, nanomedicine, and immune-oncology deepens, these systems are likely to become increasingly customized, combinatorial, and intelligent, heralding a new era of precision medicine for ovarian cancer treatment.
5. Liposomes in Cancer Therapy Beyond Ovarian Cancer:
5.1. Pegylated Liposomal Doxorubicin (Doxil/Caelyx):
Approved in 1995, PLD remains the archetype. By remote?loading doxorubicin into PEGylated vesicles (~100?nm), PLD achieves a plasma half?life of 55?hours and ten?fold higher tumor exposure compared with free drug [12].
Pegylated liposomal doxorubicin (PLD), marketed as Doxil® (US) and Caelyx® (EU/Canada), is a landmark nanomedicine formulation widely used in oncology beyond ovarian cancer. Encapsulation of doxorubicin in PEGylated liposomes extends its circulation half-life, enhances tumor accumulation via the enhanced permeability and retention (EPR) effect, and reduces cardiotoxicity a major dose-limiting toxicity of free doxorubicin.
PLD is approved as a monotherapy for patients with MBC, especially those with cardiac risk factors or who have failed prior anthracycline treatment. It is preferred due to its lower cardiotoxicity profile. In Phase III trials, PLD showed comparable efficacy to conventional doxorubicin with significantly reduced heart failure rates [65].
In combination with bortezomib, PLD improves progression-free survival in relapsed/refractory multiple myeloma. This combination enhances anti-myeloma activity without adding significant toxicity [86].
PLD remains a first-line treatment for AIDS-related Kaposi’s Sarcoma. Its efficacy, safety, and outpatient convenience have made it the standard of care since the late 1990s [87].
PLD has been tested in advanced/recurrent endometrial cancers with modest activity. It is sometimes used off-label when platinum-based regimens fail [88].
While not FDA-approved for HCC, PLD has been explored in Phase II studies due to its altered pharmacokinetics and safer hepatic profile [89].
PEGylation shields the liposome, avoiding rapid clearance and minimizing free drug exposure to the heart.
PLD exhibits a circulation half-life of ~55 hours vs. ~20 minutes for free doxorubicin.
Exploits the EPR effect for better tumor uptake.
Less systemic exposure results in a milder side-effect profile.
A unique dose-limiting side effect of PLD due to accumulation in skin capillaries.
Though rare, PLD can cause hypersensitivity reactions requiring premedication.
The formulation is significantly more expensive than generic doxorubicin.
5.2. Vyxeos (CPX?351):
Vyxeos encapsulates a synergistic 5:1 molar ratio of daunorubicin to cytarabine, demonstrating that liposomes can co?load fixed drug ratios unachievable by co?infusion. Vyxeos® (CPX?351) is a dual-drug liposomal formulation co-encapsulating daunorubicin and cytarabine in a fixed, synergistic molar ratio of 1:5. Approved by the FDA in 2017 and subsequently by the EMA, Vyxeos represents a major advancement in liposomal nanomedicine, specifically for the treatment of high-risk acute myeloid leukemia (AML) a malignancy that has historically posed treatment challenges due to poor prognosis and the toxicity of intensive chemotherapy.
5.2.1. Mechanism and Design Innovation
Vyxeos uses a bilamellar liposomal vesicle system to co-encapsulate daunorubicin and cytarabine at a fixed 1:5 molar ratio, enabling:
This fixed ratio was determined through preclinical synergy screening, maximizing anti-leukemic activity while reducing drug antagonism between the two agents [90].
5.2.2. Indications Beyond Ovarian Cancer
Vyxeos has clinical application primarily in hematologic malignancies:
1. Therapy-Related AML (t?AML)
2. AML with Myelodysplasia-Related Changes (AML?MRC)
3. Potential Expansion (Investigational)
5.2.3. Clinical Efficacy
Table No. 4: A pivotal Phase III trial compared Vyxeos to conventional 7+3 chemotherapy in older adults with newly diagnosed high-risk AML [91]
Outcome |
Vyxeos |
7+3 Standard Therapy |
Median Overall Survival |
9.56 months |
5.95 months |
Complete Remission (CR) |
37.3% |
25.6% |
60-day Mortality |
13.7% |
21.2% |
Time to Transplant |
Shorter |
Longer |
These findings led to its regulatory approvals and established Vyxeos as standard of care in high-risk AML subtypes.
5.2.4. Advantages Over Conventional Chemotherapy
5.2.5. Limitations and Toxicities
5.3. Onivyde (Liposomal Irinotecan):
Onivyde’s sucrose?octasulfate gradient stabilizes irinotecan in its active lactone form, extending half?life and reducing gastrointestinal toxicity in pancreatic cancer. Onivyde® (also known as nal-IRI or liposomal irinotecan) is a liposomal formulation of the topoisomerase I inhibitor irinotecan, designed to improve drug delivery and pharmacokinetics in solid tumors, particularly pancreatic cancer. It was approved by the FDA in 2015 for use in combination therapy for metastatic pancreatic adenocarcinoma following gemcitabine-based treatment.
5.3.1. Mechanism and Liposomal Design
Onivyde encapsulates irinotecan in a PEGylated liposomal carrier, which:
PEGylation also contributes to the stability of the liposome and reduces opsonization and clearance by the mononuclear phagocyte system (MPS) [73].
5.3.2. Approved Clinical Indication
Metastatic Pancreatic Cancer: Onivyde is FDA-approved for use in combination with fluorouracil (5-FU) and leucovorin in patients with metastatic pancreatic cancer who have progressed on gemcitabine-based therapy. Pancreatic cancer typically has a poor prognosis, and Onivyde has demonstrated a survival benefit in this difficult-to-treat population.
5.3.3. Clinical Efficacy (NAPOLI-1 Trial)
In the Phase III NAPOLI-1 trial, patients with metastatic pancreatic cancer previously treated with gemcitabine were randomized to receive either:
Table No. 5: Key outcomes of Clinical Efficacy (NAPOLI-1 Trial) [93]
Outcome |
Onivyde Combo |
5-FU/Leucovorin Alone |
Median Overall Survival |
6.1 months |
4.2 months |
Progression-Free Survival |
3.1 months |
1.5 months |
Objective Response Rate |
16% |
1% |
These results led to regulatory approval and support its use as second-line therapy in pancreatic cancer.
5.3.4. Potential Beyond Pancreatic Cancer (Investigational)
Although Onivyde is currently approved for pancreatic cancer, clinical trials have investigated or are ongoing in:
Liposomal irinotecan demonstrates enhanced intratumoral SN-38 levels in several tumor types, suggesting broader applicability [94].
5.3.5. Advantages Over Conventional Irinotecan
5.3.6. Limitations and Toxicity
6. Clinically Approved and Investigational Liposomal Therapies in Ovarian Cancer:
6.1. Pegylated Liposomal Doxorubicin (PLD):
A 2024 meta?analysis of seven randomized trials (n?=?3?812) demonstrated that PLD?carboplatin significantly prolonged progression?free survival versus paclitaxel?carboplatin (hazard ratio 0.88) [13]. In platinum?resistant disease, PLD monotherapy yields an objective response rate of around 20?%.
Pegylated Liposomal Doxorubicin (PLD)
Pegylated Liposomal Doxorubicin (PLD), commercially known as Doxil® (US) or Caelyx® (EU), is a landmark in nanomedicine and a first-line liposomal chemotherapeutic agent approved for the treatment of platinum-resistant ovarian cancer. It represents one of the most clinically validated liposomal formulations in oncology.
PLD is approved for:
Numerous clinical trials have demonstrated the efficacy and safety of PLD in ovarian cancer.
Table No. 6: Advantages of PLD Over Free Doxorubicin
Feature |
Free Doxorubicin |
PLD |
Circulation time |
Short |
Prolonged (PEGylation) |
Cardiotoxicity |
High |
Reduced |
Tumor accumulation |
Non-specific |
Enhanced (via EPR effect) |
Alopecia & mucositis |
Common |
Less frequent |
Dosing frequency |
More frequent |
Once every 4 weeks |
While PLD has improved outcomes in ovarian cancer, resistance and limited penetration in poorly vascularized tumor areas remain challenges. Future iterations include:
6.2. Lipoplatin:
Lipoplatin incorporates cisplatin into 110?nm vesicles to reduce nephrotoxicity. A phase?II trial reported grade 3?4 nephrotoxicity in none of the lipoplatin?treated patients versus 14?% with free cisplatin [14]. Lipoplatin is a liposomal encapsulation of the widely used chemotherapeutic agent cisplatin, developed to improve the therapeutic index of the drug by reducing systemic toxicity especially nephrotoxicity and enhancing tumor accumulation via the enhanced permeability and retention (EPR) effect [69,70].
6.2.1. Composition and Structure
Lipoplatin consists of cisplatin encapsulated in a liposome made from dipalmitoyl phosphatidyl glycerol (DPPG), soy phosphatidylcholine (SPC), cholesterol, and methoxy-polyethylene glycol-distearoylphosphatidylethanolamine (mPEG-DSPE). These components form a PEGylated liposome, which prolongs circulation time in the bloodstream and facilitates better tumor targeting [69,71].
6.2.2. Mechanism of Action
Lipoplatin acts via:
6.2.3. Preclinical and Clinical Investigations in Ovarian Cancer
Preclinical models have demonstrated increased accumulation of Lipoplatin in tumor tissues compared to free cisplatin [70,71]. Phase I/II trials have shown favorable safety profiles, particularly reduced renal and gastrointestinal toxicity [69]. In ovarian cancer, Lipoplatin has been explored both as a monotherapy and in combination with other agents like paclitaxel [72].
One clinical study found that the combination of Lipoplatin with paclitaxel yielded similar efficacy to standard cisplatin-paclitaxel regimens but with significantly lower nephrotoxicity and better patient tolerability [72].
6.2.4. Advantages over Conventional Cisplatin
6.2.5. Limitations and Challenges
6.2.6. Current Status
Lipoplatin has shown promising results in Phase II and III trials in various cancers (notably NSCLC), and though not yet widely approved for ovarian cancer, it remains an active area of research for platinum-resistant and recurrent disease cases [69,72].
6.3. OSI?211 (Liposomal Lurtotecan):
Early?phase trials showed modest response rates; modern ionizable lipid formulations are under re?evaluation. OSI?211, also known as Liposomal Lurtotecan, is an investigational liposomal formulation of lurtotecan, a water-insoluble camptothecin analogue and a topoisomerase-I inhibitor. Developed to address the challenges of drug solubility, toxicity, and limited efficacy of conventional camptothecins, OSI?211 has been explored as a potential therapy for platinum-resistant ovarian cancer.
6.3.1. Formulation and Design Rationale
Lurtotecan, the active agent, is encapsulated within a sterically stabilized liposome that contains:
This composition enhances the drug's pharmacokinetics, extends its plasma half-life, and enables passive tumor targeting via the enhanced permeability and retention (EPR) effect [73,74].
6.3.2. Mechanism of Action
Lurtotecan inhibits DNA topoisomerase I, an enzyme critical for DNA replication and transcription. By stabilizing the enzyme-DNA cleavable complex, it causes DNA strand breaks and induces cell death in rapidly dividing tumor cells [74].
The liposomal delivery system:
6.3.3. Clinical Investigation in Ovarian Cancer
In Phase II clinical trials, OSI?211 was administered intravenously to patients with platinum- and paclitaxel-resistant ovarian cancer. The studies reported:
Despite encouraging pharmacokinetics and safety, the efficacy results were not sufficient to support further development for ovarian cancer, and development was discontinued [75,76].
6.3.4. Advantages of OSI?211
6.3.5. Limitations
6.3.6. Current Status
While OSI?211 is not approved and has been discontinued in ovarian cancer trials, it remains an important example of how liposomal encapsulation can enhance the delivery of otherwise challenging chemotherapeutic agents.
6.4. Combination Liposomes:
Folate?receptor?targeted liposomes co?encapsulating niraparib and doxorubicin achieved synergistic cytotoxicity and 85?% tumor regression in orthotopic models [15]. A first?in?human dose?escalation study began in 2024.
Combination liposomes are an advanced class of liposomal drug delivery systems engineered to co-encapsulate two or more therapeutic agents within a single nanocarrier. These systems aim to enhance synergistic anticancer effects, optimize drug ratios, and reduce systemic toxicity, particularly for multidrug-resistant and recurrent ovarian cancer.
6.4.1. Rationale for Combination Liposomal Therapy
Ovarian cancer frequently requires combination chemotherapy due to:
Traditional combination regimens often suffer from inconsistent pharmacokinetics and differential toxicity of the co-administered drugs. Liposomal co-delivery offers a solution by synchronizing the delivery and biodistribution of multiple drugs [6,77].
6.4.2. Design and Mechanism
Combination liposomes are typically constructed using:
Once delivered to the tumor site via the enhanced permeability and retention (EPR) effect, the liposome releases both drugs in a controlled fashion to maintain the synergistic ratio at the cellular level [77,73].
6.4.3. Key Examples in Ovarian Cancer Research
6.4.4. Advantages Over Single-Agent Liposomes
6.4.5. Challenges and Limitations
6.4.6. Current Status and Future Prospects
Although combination liposomes have not yet received clinical approval for ovarian cancer, several formulations are in preclinical and early-phase trials. Their ability to address drug resistance and improve response rates makes them a promising direction in next-generation nanomedicine for ovarian cancer therapy.
6.5. TTFields?Compatible Liposomes:
Tumor?Treating Fields intensify membrane permeability, enhancing liposome uptake. Interim analysis of the INNOVATE?3 study suggests a potential survival benefit [16].
6.5.1. Tumor Treating Fields (TTFields) are an emerging, non-invasive cancer therapy that employs low-intensity, intermediate-frequency alternating electric fields to disrupt cancer cell division. In ovarian cancer, TTFields have shown potential when combined with conventional chemotherapy, particularly liposomal drug delivery systems, to enhance therapeutic outcomes while minimizing systemic toxicity [80].
6.5.2. TTFields-compatible liposomes are designed to retain their stability and function under the influence of alternating electric fields. These nanocarriers offer a dual approach leveraging the cytotoxic effects of both the encapsulated chemotherapeutic agents and the mitosis-disrupting action of TTFields [81]. Preclinical studies have demonstrated that TTFields can increase the permeability of cancer cell membranes, thereby enhancing the uptake of liposomes and improving drug delivery efficiency [82].
In addition, TTFields may facilitate localized and enhanced drug release from liposomes due to mechanical stress and changes in membrane dynamics. This results in increased intracellular drug concentration and greater cancer cell apoptosis [83].
6.5.3. Design of TTFields-Compatible Liposomes
To ensure effective co-delivery with TTFields, liposomes must meet certain structural criteria:
6.5.4. Investigational Use in Ovarian Cancer
Although TTFields-compatible liposomes are still in the investigational phase for ovarian cancer, combination therapies using pegylated liposomal doxorubicin (PLD) and TTFields have shown synergistic effects in both in vitro and in vivo models. These studies indicate higher drug accumulation at tumor sites and significantly greater tumor growth inhibition than either therapy alone [85].
This combination strategy holds promise for overcoming drug resistance in ovarian cancer, especially in recurrent or platinum-resistant cases. The integration of bioelectronic TTFields with nanotechnology-based delivery systems represents a novel therapeutic frontier.
7. Preclinical Advances: Gene, RNA and Immune Modulation:
7.1. Gene Therapy Vectors
A 2023 systematic review of murine studies confirmed that cationic liposome?mediated gene delivery reduced mean tumor weight by 63?% and extended median survival by 25 days [17].
7.2. microRNA?Loaded Liposomes
microRNA?7 lipoplexes target EGFR and impair spheroid formation, while multiplex loading of miR?200c and miR?199a?3p is being explored to tackle epithelial–mesenchymal transition and angiogenesis [18].
7.3. Immuno?Liposomes and Oncolytic Viruses
Cationic liposomes formulated with DOTAP/DOPE facilitate reovirus entry, enhancing ovarian cancer cell kill in vitro by three?fold [19]. CD47?blocking immuno?liposomes improve phagocytic clearance of cancer spheroids.
7.4. Exosome?Mimetic Liposomes
Hybrid exosome?liposome nanovesicles combine innate tropism with scalable production, doubling peritoneal retention and reversing paclitaxel resistance in SKOV3?TR cells [20].
8. Emerging Designer Liposomes:
8.1. Stimuli?Responsive Platforms
Endogenous stimuli such as acidic pH and elevated glutathione trigger selective payload release. A 2025 review catalogued more than 80 pH?sensitive systems [21].
Stimuli-responsive liposomes also called smart or triggered liposomes are advanced nanocarriers engineered to release their therapeutic payload in response to specific external or internal stimuli. These designer systems aim to enhance target specificity, minimize off-target toxicity, and overcome biological barriers, especially in solid tumours such as ovarian cancer.
8.1.1. Types of Stimuli and Mechanisms
a) pH-Responsive Liposomes
b) Temperature-Responsive Liposomes
c) Enzyme-Responsive Liposomes
d) Redox-Responsive Liposomes
e) Light-Responsive Liposomes
8.2. Ligand?Targeted Approaches:
Beyond folate receptors, transferrin, integrin αvβ3, and EphA2 antibodies furnish targeting moieties, substantially increasing tumoral uptake without raising hepatic burden.
Ligand-targeted liposomes represent a powerful strategy in the design of “smart” or designer liposomes, aiming for active targeting of specific cell types especially cancer cells by using ligands that bind to overexpressed receptors on the target cell surface. This enhances cellular uptake, minimizes off-target effects, and improves therapeutic efficacy.
8.2.1. Mechanism of Action
8.2.2. Common Ligands Used
a. Monoclonal Antibodies (Immunoliposomes)
b. Peptides
c. Aptamers
d. Folate
8.2.3. Advantages
8.2.4. Challenges
8.2.5. Emerging Trends
8.2.6. Applications in Ovarian Cancer
8.3. Multimodal and Imageable Liposomes:
Gadolinium?doped bilayers enable real?time MRI mapping; 64Cu?chelated lipids provide PET tracking and dosimetry.
Multimodal and imageable liposomes are an advanced class of liposomal nanocarriers that combine therapeutic and diagnostic capabilities a concept referred to as theranostics. These platforms enable real-time tracking of biodistribution, monitoring of drug delivery, and simultaneous treatment and imaging, offering immense value in personalized and precision medicine.
8.3.1. Multimodal Liposomes
These liposomes are engineered to combine multiple functionalities within a single vesicle, such as:
Such platforms support targeted therapy, controlled release, and simultaneous visualization of drug delivery and therapeutic response [42].
8.3.2. Imageable Liposomes
These liposomes are designed to carry imaging agents that allow non-invasive detection and monitoring via medical imaging technologies.
A. Magnetic Resonance Imaging (MRI)
B. Fluorescence Imaging
C. Computed Tomography (CT)
D. Positron Emission Tomography (PET) / Single Photon Emission CT (SPECT)
8.3.4. Applications in Cancer Therapy
8.3.5. Advantages
8.3.6. Limitations
8.3.7. Future Directions
8.4. Sonodynamic and Photodynamic Hybrids:
Porphyrin?lipid conjugates form 'porphysomes' that generate singlet oxygen upon irradiation, offering combined therapeutic and imaging capabilities [22].
Sonodynamic and photodynamic liposomes represent a frontier in non-invasive cancer therapy, combining light or ultrasound-based activation with liposomal drug delivery. These hybrid systems incorporate photosensitizers or sonosensitizers into the liposome, enabling controlled release and cytotoxicity upon external stimulation.
Such systems not only localize drug release but also generate reactive oxygen species (ROS) that induce tumour cell apoptosis, offering synergistic antitumor effects while minimizing damage to surrounding healthy tissue.
8.4.1. Photodynamic Liposomes (PDT-Liposomes)
a. Mechanism:
b. Design Features:
8.4.2. Sonodynamic Liposomes (SDT-Liposomes)
a. Mechanism:
b. Design Features:
8.4.3. Advantages
8.4.4. Challenges
8.4.5. Applications in Cancer Therapy
8.4.6. Future Outlook
9. Translational Barriers and Knowledge Gaps:
EPR heterogeneity, accelerated blood clearance in patients with pre?existing anti?PEG antibodies, immunogenic toxicity, and manufacturing scalability remain principal obstacles [23]. Regulatory authorities now emphasize lipid impurity profiling and thermal stability assessments [24].
10. Future Perspectives:
Key trends include precision nanomedicine, combinatorial therapeutics, intraperitoneal smart delivery, and AI?assisted formulation. Integrating genomic data with quantitative imaging may enable patient?tailored dosing.
CONCLUSION:
Liposome technology has profoundly transformed the landscape of drug delivery, particularly in the context of ovarian cancer a malignancy characterized by late diagnosis, peritoneal spread, and resistance to conventional chemotherapies. Over the past three decades, innovations in liposome composition, targeting strategies, and responsive release mechanisms have enabled the delivery of cytotoxic agents, immunomodulators, and genetic material with heightened specificity and reduced systemic toxicity.
Pegylated liposomal doxorubicin (PLD) set a precedent for nanocarrier-based chemotherapeutics, offering a favourable safety profile and improved pharmacokinetics. Subsequent developments such as folate-receptor-targeted liposomes, gene-loaded vectors, and exosome-mimetic systems demonstrate the versatility of liposomes in overcoming biological barriers, enhancing tumour selectivity, and reprogramming the tumour microenvironment. Preclinical and clinical evidence supports the superiority of these platforms in improving therapeutic index, mitigating adverse effects, and extending survival particularly in platinum-resistant or recurrent ovarian cancer.
Nonetheless, significant translational barriers persist. The heterogeneity of the enhanced permeability and retention (EPR) effect, immune clearance mechanisms like anti-PEG antibodies, and manufacturing challenges demand continued refinement. Regulatory frameworks must evolve in parallel with technological advances to ensure safety, reproducibility, and scalability.
Future directions point toward precision nanomedicine, where liposomal formulations are matched to tumour genotype, microenvironmental cues, and patient-specific pharmacodynamics. The integration of real-time imaging, AI-guided formulation design, and combinatorial payloads holds promise for individualized, adaptive cancer therapy. As liposome science enters its fourth decade, its role in ovarian cancer therapy is no longer adjunctive but central ushering in a new era of intelligent, programmable therapeutics that align closely with the goals of modern oncology.
Liposomes stand at the forefront of nanomedicine for ovarian cancer. Three decades of clinical experience with PLD validate their safety and efficacy, while targeted, stimuli?responsive, and gene?loaded designs promise to address persistent unmet needs such as platinum resistance and peritoneal metastasis.
ACKNOWLEDGEMENT:
This work is part of first year M. Pharmacy (Pharmaceutics) assignment of first author. The authors gratefully acknowledge the guide, Ms. Vidya Thorat, Dr. P. N. Sable and special gratitude to Dr. Rupali H. Tiple assisting for the successful completion of the project.
REFERENCES
Kiran Naik, Vidya Thorat, P. N. Sable, Rupali Tiple, Liposomes in the Treatment of Ovarian Cancer, and Liposomal Therapeutics: A Comprehensive Review, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 8, 1985-2015. https://doi.org/10.5281/zenodo.16903990