View Article

  • Role Of Lipid Carriers in Overcoming Drug Resistance in Cancer

  • Department of Pharmaceutics, MB School of Pharmaceutical Sciences, Mohan Babu University, Sree Sainath Nagar, Tirupati 517102

Abstract

Drug resistance remains a significant challenge in cancer therapy, leading to treatment failure and disease progression. This resistance, caused by mechanisms such as efflux transporter overexpression, altered drug targets, and anti-apoptotic pathways, limits the efficacy of conventional chemotherapeutics. Lipid carriers, including liposomes, solid lipid nanoparticles (SLNs), and nanostructured lipid carriers (NLCs), have emerged as promising systems to address these challenges. Lipid carriers offer advantages such as improved drug solubility, stability, enhanced permeability, targeted delivery, and controlled release. Furthermore, they can circumvent resistance mechanisms like P-glycoprotein (P-gp)-mediated efflux and improve intracellular drug accumulation through endocytosis-based uptake. By enabling co-delivery of chemotherapeutic agents and resistance pathway inhibitors, lipid carriers provide a multifunctional approach to overcome multidrug resistance. This article provides a detailed overview of the mechanisms of drug resistance in cancer, the advantages of lipid carriers, their specific strategies for addressing resistance pathways, and their role in enhancing the therapeutic efficacy of chemotherapeutic agents.

Keywords

Drug resistance, Lipid carriers, Cancer therapy, Efflux transporters, Targeted drug delivery.

Introduction

Overview of cancer treatment challenges

Cancer remains one of the most complex diseases to treat due to its heterogeneity, genetic instability, and adaptability to therapeutic interventions. Traditional cancer therapies, such as chemotherapy, radiotherapy, and surgery, often fail to achieve complete remission because of their inability to distinguish between healthy and cancerous cells1. This lack of specificity results in systemic toxicity, severe side effects, and dose-limiting toxicities, which hinder the effectiveness of treatment. Additionally, the tumor microenvironment (TME), which includes hypoxia, acidic pH, and immune suppression, creates a protective niche that enables cancer cells to evade therapeutic agents and promotes survival2,3.

Moreover, drug resistance further exacerbates these challenges. Tumors develop intrinsic resistance through genetic mutations and adaptive resistance after repeated exposure to chemotherapeutics. This phenomenon is characterized by reduced drug accumulation, enhanced DNA repair mechanisms, and upregulation of anti-apoptotic pathways. Furthermore, overexpression of efflux transporters, such as P-glycoprotein, significantly lowers intracellular drug concentrations, reducing treatment efficacy. As a result, there is a growing need for advanced drug delivery systems that can target tumors efficiently, enhance drug uptake, and overcome resistance mechanisms4,5.

Importance of addressing drug resistance mechanisms

Drug resistance in cancer treatment is a major cause of therapeutic failure, resulting in high mortality and relapse rates among patients. Resistance mechanisms can either be intrinsic (pre-existing) or acquired after exposure to chemotherapeutics. Intrinsic resistance occurs due to the inherent genetic instability of cancer cells, which allows them to survive and proliferate despite drug treatment6. Acquired resistance, on the other hand, develops over time as cancer cells adapt to therapeutic stress through molecular changes. Understanding and addressing these resistance mechanisms is crucial for improving cancer treatment outcomes7,8.

One of the most prominent mechanisms of drug resistance involves the overexpression of efflux transporters, particularly ATP-binding cassette (ABC) transporters like P-glycoprotein (P-gp), multidrug resistance-associated proteins (MRPs), and breast cancer resistance protein (BCRP). These transporters actively pump chemotherapeutic agents out of cancer cells, reducing intracellular drug concentration and efficacy. In addition, tumor cells can alter drug targets, such as topoisomerase or tubulin, making them less susceptible to drug binding9. Overcoming such mechanisms requires advanced delivery systems that can bypass efflux pumps and deliver sufficient drug concentrations directly to target cells.

Addressing drug resistance is further complicated by the tumor microenvironment (TME), which provides protective conditions for cancer cells. The TME is characterized by hypoxia, acidic pH, and elevated levels of growth factors that promote cancer cell survival, metastasis, and resistance. Moreover, cancer cells activate anti-apoptotic pathways and enhance their DNA repair mechanisms to evade the effects of chemotherapeutic agents. By targeting these resistance mechanisms through innovative approaches like lipid carriers, therapeutic agents can be delivered effectively to cancer cells while minimizing systemic toxicity10. Lipid carriers offer the advantage of targeted delivery, controlled release, and co-delivery of drugs and resistance-modulating agents, making them an ideal solution to address these challenges.

The importance of addressing drug resistance cannot be overstated, as it directly impacts patient survival, treatment efficacy, and quality of life. Developing novel strategies to overcome resistance mechanisms, such as lipid-based nanocarriers, can significantly improve therapeutic outcomes. These systems not only enhance the pharmacokinetics and bioavailability of drugs but also provide opportunities to inhibit resistance pathways, such as efflux transporters and anti-apoptotic proteins11. Future advancements in lipid carrier technologies, including functionalization and stimuli-responsive systems, hold promise for overcoming drug resistance and achieving better clinical success in cancer therapy.

Figure- Overcoming Cancer Strategies by Lipid Formulations

Mechanisms of Drug Resistance in Cancer

The mechanisms of drug resistance is essential for designing effective therapies. The key mechanisms include:

Role of ATP-binding cassette (ABC) transporters in drug efflux

ATP-binding cassette (ABC) transporters are integral membrane proteins that utilize ATP hydrolysis to translocate a variety of substrates across cellular membranes, playing a pivotal role in drug efflux mechanisms. Recent studies have provided deeper insights into their structural and functional dynamics, enhancing our understanding of their role in pharmacokinetics and multidrug resistance12.

Advancements in structural biology, particularly through cryo-electron microscopy, have elucidated the conformational changes ABC transporters undergo during substrate translocation. These findings have been complemented by computational modeling, offering a comprehensive view of the transport mechanisms and aiding in the development of potential inhibitors to modulate their activity13,14.

In the context of the blood-brain barrier (BBB), specific ABC transporters such as P-glycoprotein (P-gp), breast cancer resistance protein (BCRP), and multidrug resistance protein 4 (MRP4) have been identified as key players in limiting the central nervous system (CNS) penetration of antiviral agents like ganciclovir. Inhibition of these transporters has been shown to enhance drug permeability across the BBB, suggesting a potential strategy to improve therapeutic efficacy in CNS infections15.

The role of ABC transporters extends beyond drug efflux; they are also involved in maintaining cellular metabolic fitness. For instance, MDR1 (P-gp) has been implicated in regulating oxidative stress responses in activated T cells, indicating its significance in immune cell function and potential implications in immunotherapy. In cancer therapy, the efficacy of treatments like photodynamic therapy (PDT) can be compromised by ABC transporter-mediated efflux of photosensitizers. Studies assessing the susceptibility of various photosensitizers to transport by P-gp, BCRP, and MRP1 have highlighted the need to consider transporter interactions to enhance PDT outcomes. Furthermore, the interplay between ion channels and the PI3K/AKT signaling pathway has been linked to the regulation of ABC transporter expression, contributing to chemoresistance. Understanding this relationship opens avenues for targeting ion channels as a therapeutic strategy to modulate ABC transporter activity and overcome drug resistance in cancer cells12,15,16.

Mechanisms of P-glycoprotein (P-gp) overexpression and its impact on drug efficacy

P-glycoprotein (P-gp), encoded by the ABCB1 gene, is an ATP-dependent efflux transporter that plays a significant role in multidrug resistance (MDR) by expelling a wide range of chemotherapeutic agents from cancer cells, thereby reducing their efficacy. Overexpression of P-gp in tumor cells is a major obstacle in cancer treatment, as it leads to decreased intracellular drug accumulation and diminished therapeutic outcomes17.

Table-1: Mechanisms of Drug Resistance in Cancer

Mechanism

Description

Examples

References

Efflux Transporters55

Membrane proteins actively pump chemotherapeutics out of cancer cells, lowering intracellular drug concentration.

P-gp, BCRP, MRP1-mediated drug efflux

Gottesman et al., 2016

Drug Target Alterations56

Cancer cells mutate or alter drug-binding sites, reducing the efficacy of targeted therapies.

Mutations in EGFR, topoisomerase, tubulin-binding proteins

Hanahan & Weinberg, 2011

Enhanced DNA Repair57

DNA repair enzymes counteract drug-induced DNA damage, preventing apoptosis.

BRCA1/2 mutations enhancing DNA repair

Longley & Johnston, 2005

Anti-apoptotic Pathways58

Upregulation of survival proteins like Bcl-2 and activation of PI3K/Akt pathways lead to resistance.

Overexpression of Bcl-2, Akt, NF-κB activation

Zahreddine & Borden, 2013

Epithelial-Mesenchymal Transition (EMT)59

Cancer cells gain mesenchymal-like traits, increasing invasion, metastasis, and drug resistance.

Upregulation of Snail, Twist, and Zeb1 proteins

Junttila & de Sauvage, 2013

Tumor Microenvironment (TME)60

Tumor-associated factors like hypoxia, acidic pH, and immune evasion protect cancer cells from drugs.

VEGF-mediated angiogenesis, CAF interactions, acidic microenvironment

Chen et al., 2016

Autophagy-Mediated Drug Resistance61

Cancer cells use autophagy as a survival mechanism to evade chemotherapy-induced cell death.

Upregulated Beclin-1 and LC3-II promoting survival

Owais et al., 1995

Epigenetic Modifications62

DNA methylation, histone modifications, and non-coding RNAs regulate genes responsible for drug sensitivity.

Hypermethylation of tumor suppressor genes, altered miRNA expression

Patel & Patel, 2017

Impact on Drug Efficacy:

The overexpression of P-gp in cancer cells leads to the efflux of a broad spectrum of chemotherapeutic agents, including anthracyclines, taxanes, and vinca alkaloids, reducing their intracellular concentrations and therapeutic effectiveness. This efflux activity is a key mechanism behind the development of MDR in human cancers18.

Additionally, P-gp overexpression affects the pharmacokinetics of administered drugs by altering their absorption, distribution, metabolism, and excretion. For example, increased P-gp activity in the intestinal epithelium can reduce oral bioavailability of substrate drugs, while its presence in the blood-brain barrier limits central nervous system penetration, impacting the efficacy of treatments for brain tumors18.

Understanding the mechanisms underlying P-gp overexpression and its impact on drug efficacy is crucial for developing strategies to overcome MDR in cancer therapy. Approaches such as the use of P-gp inhibitors, modulation of regulatory pathways, and personalized medicine based on genetic and epigenetic profiles are being explored to enhance the effectiveness of chemotherapeutic agents in resistant cancers.

Overview of Lipid-Based Drug Delivery Systems

Liposomes

Liposomes have advanced significantly from their initial design, incorporating cutting-edge modifications that enhance functionality and expand their applications. Modern liposomes are now engineered with stimuli-responsive capabilities, enabling precise control over drug release. For instance, thermo-sensitive liposomes release their payload at elevated temperatures, ideal for use in localized hyperthermia treatments, while pH-sensitive liposomes exploit the acidic tumor microenvironment for targeted drug delivery19-21. Multifunctional liposomes, often referred to as theranostic liposomes, integrate therapeutic and diagnostic agents into a single system, allowing simultaneous treatment and real-time monitoring of disease progression. Furthermore, surface modifications with ligands such as aptamers, peptides, and antibodies have revolutionized active targeting, ensuring that liposomes bind selectively to receptors overexpressed in diseased tissues, such as HER2 in breast cancer or CD44 in metastatic cancers20-23.

Recent breakthroughs also focus on improving liposome stability and circulation time through PEGylation and incorporating cholesterol derivatives, which resist premature degradation. Additionally, hybrid liposomal systems, which combine liposomes with other nanostructures like dendrimers or gold nanoparticles, have emerged, offering enhanced drug loading and imaging capabilities. In gene therapy, cationic liposomes are optimized to encapsulate CRISPR-Cas9 complexes or siRNA, ensuring efficient delivery and gene-editing precision24,25. These innovations not only address traditional limitations such as drug leakage and short shelf life but also position liposomes as a cornerstone in personalized medicine, capable of addressing the intricacies of complex diseases with unprecedented specificity and efficacy.

Solid Lipid Nanoparticles (SLNs)

Solid Lipid Nanoparticles (SLNs) represent an advanced drug delivery platform composed of solid lipids that remain in a crystalline state at room and body temperatures, encapsulating therapeutic agents within their hydrophobic matrix. Unlike conventional carriers, SLNs are typically made from biocompatible lipids such as glyceryl monostearate, stearic acid, or triglycerides, stabilized by surfactants like polysorbates or lecithins26,27. Recent innovations have focused on optimizing the lipid matrix to enhance drug loading efficiency and reduce polymorphic transitions, which can affect stability28. Furthermore, the use of hybrid stabilizers, such as polymer-lipid conjugates, has significantly extended the shelf life of SLNs by preventing particle aggregation and maintaining their colloidal stability over time.

In drug delivery, SLNs offer unique advantages, including the ability to enhance the bioavailability of poorly water-soluble drugs, protect labile drugs from degradation, and provide controlled or sustained release profiles. Advanced applications leverage SLNs for site-specific delivery through surface functionalization with targeting ligands, such as folic acid or antibodies, enabling precise interaction with diseased tissues29. SLNs are particularly impactful in cancer therapy, where they improve the solubility and efficacy of chemotherapeutics like paclitaxel, while reducing systemic toxicity. Additionally, their potential in gene delivery has been realized through the encapsulation of nucleic acids, such as siRNA and DNA, in cationic lipid matrices. The ability to co-deliver multiple therapeutic agents and incorporate diagnostic imaging agents positions SLNs as versatile and promising candidates in the emerging field of theranostics, offering an integrated approach to treatment and monitoring30.

Nanostructured Lipid Carriers (NLCs)

Nanostructured Lipid Carriers (NLCs) are a second-generation lipid-based drug delivery system, specifically designed to overcome the inherent limitations of Solid Lipid Nanoparticles (SLNs). By incorporating a blend of solid and liquid lipids, NLCs achieve a less-ordered crystalline matrix, which significantly enhances their drug-loading capacity and stability31,32. This unique structural configuration prevents the expulsion of drugs during storage, a common issue in SLNs caused by lipid recrystallization. Additionally, the inclusion of liquid lipids allows for encapsulation of a broader range of therapeutic agents, including hydrophobic and hydrophilic drugs, while maintaining a controlled and sustained release profile. These properties make NLCs ideal for applications in challenging therapeutic areas, such as cancer and neurodegenerative diseases, where long-term drug efficacy is critical33.

The targeting capabilities of NLCs have been significantly advanced through functionalization strategies. Ligands such as folic acid, peptides, and antibodies can be conjugated to the NLC surface to achieve active targeting of specific cell receptors, enhancing precision drug delivery. Furthermore, stimuli-responsive NLCs, designed to release their payload in response to pH changes, temperature, or enzymatic activity, ensure localized drug delivery in diseased tissues. Beyond single-drug encapsulation, NLCs also enable the co-delivery of multiple agents, such as chemotherapeutics and siRNA, opening doors to combination therapies with synergistic effects. Their application extends to theranostics, where NLCs integrate therapeutic and diagnostic functionalities, allowing real-time tracking of drug delivery and efficacy. These advanced features position NLCs as a transformative platform in personalized medicine and next-generation drug delivery systems34,35.

Bottom of Form

Advantages of Lipid Carriers in Cancer Therapy

Enhanced Drug Solubility and Stability

Lipid carriers play a crucial role in enhancing the solubility and stability of poorly water-soluble anticancer drugs, a common challenge in cancer therapy. Hydrophobic drugs, such as paclitaxel and docetaxel, are effectively encapsulated within the lipid matrix or bilayer of carriers like solid lipid nanoparticles (SLNs), nanostructured lipid carriers (NLCs), and liposomes36. This encapsulation protects the drugs from hydrolysis, oxidation, and enzymatic degradation, thereby prolonging their stability and therapeutic efficacy. Additionally, lipid carriers improve bioavailability by promoting drug solubilization in the gastrointestinal tract or bloodstream, ensuring better absorption and systemic distribution. These properties are especially critical for drugs with low oral bioavailability or those that degrade rapidly in the physiological environment. The ability of lipid carriers to preserve the integrity and activity of anticancer agents significantly contributes to their clinical success37.

Targeted and Controlled Drug Delivery

Lipid carriers offer significant advantages in targeted and controlled drug delivery through surface functionalization and ligand-targeting strategies. By attaching ligands such as folic acid, transferrin, or antibodies to the lipid carrier surface, the system can selectively bind to overexpressed receptors on cancer cells, such as folate receptors or HER2. This ligand-receptor interaction ensures that the drug accumulates preferentially in the tumor tissue, sparing healthy cells from off-target effects36,38. Furthermore, lipid carriers provide controlled and sustained drug release due to their optimized matrix design, which maintains therapeutic drug concentrations for extended periods. Advanced systems such as stimuli-responsive lipid carriers are engineered to release drugs in response to tumor-specific triggers, like acidic pH or elevated temperatures, further enhancing precision. These targeting and release capabilities not only improve therapeutic outcomes but also reduce the required drug doses, minimizing side effects39.

Overcoming Systemic Toxicity

Systemic toxicity is a major limitation of conventional chemotherapeutic agents, which often affect both cancerous and normal cells. Lipid carriers address this issue by encapsulating drugs within their structure, reducing their direct interaction with healthy tissues. This encapsulation minimizes off-target effects and significantly improves the therapeutic index40. For instance, liposomal formulations of doxorubicin (e.g., Doxil) have been shown to reduce cardiotoxicity by restricting the drug’s distribution to the tumor site. Additionally, lipid carriers enable localized drug delivery through passive targeting mechanisms such as the enhanced permeability and retention (EPR) effect, which exploits the leaky vasculature of tumors41-43. This selective accumulation ensures that the drug is delivered primarily to the tumor site, reducing systemic exposure. By enhancing drug delivery specificity, lipid carriers provide a safer and more effective cancer treatment modality.

Prolonged Circulation Time

The pharmacokinetics of anticancer drugs can be significantly improved using lipid carriers with prolonged circulation times, achieved through PEGylation and stealth lipid systems. PEGylation, the process of attaching polyethylene glycol (PEG) chains to the lipid surface, creates a hydrophilic shield that prevents recognition and clearance by the mononuclear phagocyte system (MPS)44. This stealth property allows lipid carriers to evade immune detection, thereby increasing their half-life and ensuring sustained systemic circulation. Prolonged circulation enhances the likelihood of the carrier reaching tumor tissue via passive targeting mechanisms, such as the EPR effect. Additionally, advanced lipid carriers utilize flexible stealth coatings and tailored lipid compositions to maintain their stability and functionality in the bloodstream. These modifications not only increase drug bioavailability but also improve therapeutic outcomes by maximizing drug delivery to the tumor site while minimizing loss through rapid clearance45.

Mechanisms by Which Lipid Carriers Overcome Drug Resistance

Bypassing Efflux Transporters

Lipid carriers play a critical role in overcoming drug resistance by bypassing efflux transporters like P-glycoprotein (P-gp), which actively pump chemotherapeutics out of cancer cells, reducing intracellular drug concentrations. Unlike free drugs, lipid carriers deliver therapeutic agents via endocytosis, enabling direct cytoplasmic release and circumventing the action of membrane-bound efflux pumps. For instance, liposomal formulations encapsulate drugs within a lipid bilayer, shielding them from recognition by efflux transporters44,45. Case studies have demonstrated that lipid carriers loaded with P-gp inhibitors, such as verapamil, can enhance the efficacy of co-encapsulated chemotherapeutics like paclitaxel, overcoming multidrug resistance (MDR). Nanostructured lipid carriers (NLCs) have also been utilized to deliver siRNA targeting P-gp expression, reducing transporter activity and restoring drug sensitivity in resistant cancer cells. This dual-action approach of bypassing efflux and inhibiting transporter function exemplifies the advanced potential of lipid carriers in combating resistance46.

Enhanced Tumor Accumulation via EPR Effect

The enhanced permeability and retention (EPR) effect is a cornerstone of passive tumor targeting, and lipid carriers are uniquely designed to exploit this phenomenon. Tumor tissues possess leaky vasculature and impaired lymphatic drainage, allowing lipid nanoparticles to accumulate selectively at the site47. Additionally, active targeting strategies enhance this accumulation by functionalizing lipid carriers with ligands such as folic acid or antibodies that bind to overexpressed receptors on cancer cells. Beyond passive and active targeting, lipid carriers are engineered to respond to the tumor microenvironment's unique conditions, such as acidity or enzymatic activity48. For example, pH-sensitive lipid systems release their payload in acidic tumor tissues, ensuring localized drug delivery. Advanced formulations, like thermosensitive liposomes, respond to external hyperthermia to trigger drug release specifically at the tumor site. These strategies maximize drug concentration in tumors while minimizing systemic exposure, effectively combating resistance associated with poor drug delivery49.

Combating Altered Drug Targets

Resistance often arises from cancer cells altering or mutating drug targets, rendering conventional therapies less effective. Lipid carriers mitigate this by stabilizing chemotherapeutics and enhancing their interaction with specific targets. For example, encapsulating small molecules in liposomes or SLNs protects them from degradation and ensures their structural integrity until they reach their intended site of action50,70. Furthermore, lipid carriers enhance the bioavailability of target-specific drugs, such as tyrosine kinase inhibitors, by improving their solubility and circulation time. This is particularly beneficial in cases where resistance stems from mutations that reduce drug binding. In addition, lipid-based systems can co-deliver combination therapies, such as chemotherapeutics with allosteric inhibitors, ensuring comprehensive inhibition of both primary and altered targets. By maintaining therapeutic drug concentrations and stabilizing their active forms, lipid carriers address one of the most challenging aspects of drug resistance in cancer51.

Co-delivery of Therapeutic Agents

The co-delivery of multiple therapeutic agents using lipid carriers is a transformative approach to overcoming drug resistance. Lipid systems enable the simultaneous encapsulation and delivery of chemotherapeutics and gene therapy agents, such as siRNA or CRISPR-Cas9, allowing multi-faceted attacks on resistant cancer cells. For instance, co-delivering paclitaxel with siRNA targeting anti-apoptotic genes enhances drug efficacy by reducing cellular resistance mechanisms52. This combination approach ensures that cancer cells are sensitized to chemotherapeutics while addressing resistance at the genetic level. Additionally, co-loading two or more chemotherapeutics with complementary mechanisms of action in lipid carriers maximizes therapeutic synergy and reduces the likelihood of resistance development. These combination therapies are particularly effective against tumors with heterogenous resistance profiles, ensuring comprehensive treatment. The ability of lipid carriers to deliver agents with different physicochemical properties and release them in a controlled manner underscores their potential as a solution to multidrug resistance53.

Recent Innovations in Lipid-Based Nanocarriers

Stimuli-Responsive Lipid Carriers

Stimuli-responsive lipid carriers represent a significant advancement in drug delivery, designed to release therapeutic agents in response to specific environmental triggers. pH-sensitive carriers, for example, exploit the acidic tumor microenvironment to release drugs selectively at the tumor site, minimizing systemic toxicity49. Temperature-responsive carriers release their payload when exposed to localized hyperthermia, enhancing precision in delivery. Redox-responsive lipid carriers utilize the high glutathione levels in cancer cells to trigger drug release intracellularly. These carriers improve therapeutic efficacy by ensuring drugs are released only in target tissues, addressing challenges associated with non-specific drug distribution and resistance mechanisms53.

Functionalized and Multifunctional Lipid Carriers

Functionalized lipid carriers are engineered with surface modifications to enhance targeting and delivery efficiency. Strategies such as conjugating ligands like antibodies, peptides, or folic acid to the lipid surface allow carriers to bind specifically to overexpressed receptors on cancer cells. Multifunctional lipid carriers combine active targeting with additional features, such as stimuli-responsiveness or co-delivery capabilities, enabling personalized therapy51. For instance, PEGylation extends circulation time while simultaneously attaching targeting moieties ensures tumor-specific accumulation. These advancements allow for more precise delivery, reduced side effects, and the potential for complex therapeutic interventions, such as combination therapies53.

Theranostic Lipid Systems

Theranostic lipid systems integrate therapeutic and diagnostic functionalities into a single nanocarrier, enabling simultaneous treatment and real-time monitoring. By incorporating imaging agents such as fluorescent dyes or MRI contrast agents alongside chemotherapeutics, these carriers allow for non-invasive tracking of biodistribution and drug release. Theranostic liposomes and lipid nanoparticles have been used to deliver anticancer drugs while monitoring tumor progression via imaging. These systems enhance treatment precision by enabling clinicians to visualize the delivery process, adjust dosages in real time, and predict therapeutic outcomes, marking a significant leap toward personalized medicine and improved patient care49,54.

Table-2: Recent Innovations in Lipid-Based Drug Delivery Systems

Innovation

Description

Application

References

Stimuli-Responsive Lipid Carriers63

Lipid carriers release drugs in response to pH, temperature, enzymes, or redox conditions in tumors.

pH-sensitive liposomes, thermosensitive SLNs for tumor-specific drug release

Patel et al., 2022

PEGylation and Long-Circulating Systems64

PEGylation prolongs circulation time and reduces clearance by the immune system, enhancing accumulation in tumors.

Liposomal doxorubicin (Doxil) evading mononuclear phagocyte system

Choudhuri & Klaassen, 2006

Theranostic Lipid Nanocarriers65

Theranostic nanocarriers integrate therapeutic and diagnostic agents for real-time imaging and treatment.

MRI-visible liposomes, fluorescence-tagged NLCs for tumor imaging

Koudelka & Turánek, 2012

Co-Delivery of Multiple Therapeutic Agents66

Lipid carriers enable simultaneous delivery of chemotherapeutics, siRNA, and resistance-modulating agents.

Co-delivery of paclitaxel and siRNA targeting drug resistance genes

Rawal et al., 2021

Functionalized Lipid Carriers for Targeted Delivery67

Surface-modified lipid carriers target overexpressed receptors on cancer cells, improving specificity and efficacy.

Antibody-conjugated lipid carriers for HER2+ breast cancer therapy

Jain et al., 2015

Hybrid Nanocarriers68

Hybrid lipid-polymer or lipid-metallic systems enhance drug loading capacity and stability.

Gold nanoparticle-lipid hybrids for enhanced photothermal therapy

Mukherjee et al., 2009

CRISPR/Cas9-Loaded Lipid Systems69

Lipid nanoparticles encapsulate CRISPR/Cas9 to enable gene editing for overcoming drug resistance.

CRISPR-Cas9 lipid carriers for reversing resistance mutations

Peer et al., 2007

Artificial Intelligence (AI)-Designed Lipid Carriers71

AI-based modeling optimizes lipid formulations, improving drug loading, stability, and targeted delivery.

AI-optimized lipid carriers for personalized cancer therapy

Muthu et al., 2014

 

CONCLUSION

Lipid carriers provide a versatile platform to tackle drug resistance in cancer effectively. These systems enhance drug delivery, stability, and targeting, addressing critical resistance mechanisms. By leveraging advanced designs like stimuli-responsiveness and functionalization, lipid carriers ensure precise drug delivery to tumor sites. Co-delivery capabilities further enable synergistic therapies, overcoming multidrug resistance. Enhanced circulation time and passive targeting via the EPR effect improve drug bioavailability. Functionalized lipid carriers enable active targeting for tumor-specific accumulation. Theranostic systems integrate treatment with real-time monitoring, advancing personalized medicine. Continuous innovations in lipid carrier technologies promise better therapeutic outcomes and clinical success.

REFERENCES

  1. Gottesman, M. M., Lavi, O., Hall, M. D., & Gillet, J. P. Toward a better understanding of the complexity of cancer drug resistance. 56(1), 85–102. Annual Review of Pharmacology and Toxicology; 2016.
  2. Hanahan, D., & Weinberg, R. A. Hallmarks of cancer: The next generation. 144(5), 646–674. Cell; 2011.
  3. Junttila, M. R., & de Sauvage, F. J. Influence of tumour micro-environment heterogeneity on therapeutic response. 501(7467), 346–354. Nature; 2013.
  4. Zahreddine, H., & Borden, K. L. Mechanisms and insights into drug resistance in cancer. 4, 28. Frontiers in Pharmacology; 2013.
  5. Longley, D. B., & Johnston, P. G. Molecular mechanisms of drug resistance. 205(2), 275–292. The Journal of Pathology; 2005.
  6. Owais, M., Varshney, G. C., Choudhury, A., Chandra, S., & Gupta, C. M. Chloroquine encapsulated in malaria-infected erythrocyte-specific antibody-bearing liposomes effectively controls chloroquine-resistant Plasmodium berghei infections in mice. 39(1), 180–184. Antimicrobial Agents and Chemotherapy; 1995.
  7. Ansari, M. T., Ramlan, T. A., Jamaluddin, N. N., Zamri, N., Salfi, R., Khan, A., Sami, F., Majeed, S., & Hasnain, M. S. Lipid-based nanocarriers for cancer and tumor treatment. 26(34), 4272–4276. Current Pharmaceutical Design; 2020.
  8. Anwar, D. M., Hedeya, H. Y., Ghozlan, S. H., Ewas, B. M., & Khattab, S. N. Surface-modified lipid-based nanocarriers as a pivotal delivery approach for cancer therapy: Application and recent advances in targeted cancer treatment. 13, Article number: 106. Beni-Suef University Journal of Basic and Applied Sciences; 2024.
  9. Dechbumroong, P., Hu, R., & Liang, X.-J. Recent advanced lipid-based nanomedicines for overcoming cancer resistance. 7, 24. Cancer Drug Resistance; 2024.
  10. Benko, A., Medina-Cruz, D., & Webster, T. J. Nanocarrier drug resistant tumor interactions: Novel approaches to fight drug resistance in cancer. 4, 264–297. Cancer Drug Resistance; 2021.
  11. Chu, S., & Stochaj, U. Exploring near-infrared absorbing nanocarriers to overcome cancer drug resistance. 3, 302–333. Cancer Drug Resistance; 2020.
  12. Gottesman, M. M., & Pastan, I. Biochemistry of multidrug resistance mediated by the multidrug transporter. 62(1), 385-427. Annual Review of Biochemistry; 1993.
  13. Aller, S. G., Yu, J., Ward, A., et al. Structure of P-glycoprotein reveals a molecular basis for polyspecific drug binding. 323(5922), 1718-1722. Science; 2009.
  14. Fletcher, J. I., Haber, M., Henderson, M. J., & Norris, M. D. ABC transporters in cancer: More than just drug efflux pumps. 10(2), 147-156. Nature Reviews Cancer; 2010.
  15. Chen, Z., Shi, T., Zhang, L., et al. Mammalian drug efflux transporters of the ATP-binding cassette (ABC) family in multidrug resistance: A review of the past decade. 370(1), 153-164. Cancer Letters; 2016.
  16. Lin, J. H., & Yamazaki, M. Role of P-glycoprotein in pharmacokinetics: Clinical implications. 42(1), 59-98. Clinical Pharmacokinetics; 2003.
  17. Choudhuri, S., & Klaassen, C. D. Structure, function, expression, genomic organization, and single nucleotide polymorphisms of human ABCB1 (MDR1): Progress in understanding multidrug resistance mediated by P-glycoprotein. 34(5), 695-701. Drug Metabolism and Disposition; 2006.
  18. Seelig, A.  A general pattern for substrate recognition by P-glycoprotein. 251(1-2), 252-261. European Journal of Biochemistry; 1998.
  19. Choudhury, A., Sonowal, K., Laskar, R. E., Deka, D., & Dey, B. K. Liposome: A carrier for effective drug delivery. 10(3), 115-122. Journal of Advanced Pharmaceutical Technology & Research; 2019.
  20. Anjima, K. K., Akshaya, P., Ramalingam, N., & Vimal, K. R. Liposomal gene delivery system. 10(2), 25-32. International Journal of Pharmaceutical and Phytopharmacological Research; 2020.
  21. Agarwal, K. Liposome-assisted drug delivery: An updated review. 84(4), 322-334. Indian Journal of Pharmaceutical Sciences; 2022.
  22. Kulkarni, P. R., Yadav, J. D., & Vaidya, K. A. Liposomes: A novel drug delivery system. 3(2), 45-52. International Journal of Current Pharmaceutical Review and Research; 2011.
  23. Dhone, P. Liposomes, protein, and peptide drug delivery systems. 15(1), 12-20. International Journal of Pharmaceutical Sciences; 2024.
  24. Pawar, V., Mishra, K. S., Yadav, M., & Tiwari, A. Liposome: A powerful approach festinates drug delivery system. 3(1), 14-21. International Journal of Drug Delivery Technology; 2013.
  25. Kshirsagar, N. A., Pandya, S. K., Kirodian, B. G., & Sanath, S. Liposomal drug delivery system from laboratory to clinic. 51(3), 209-215. Journal of Postgraduate Medicine; 2005.
  26. Mehnert, W., & Mäder, K. Solid lipid nanoparticles: Production, characterization, and applications. 47(2-3), 165-196. Advanced Drug Delivery Reviews; 2001.
  27. Müller, R. H., Radtke, M., & Wissing, S. A. Solid lipid nanoparticles (SLN) and nanostructured lipid carriers (NLC) in cosmetic and dermatological preparations. 54(Suppl 1), S131-S155. Advanced Drug Delivery Reviews; 2002.
  28. Beloqui, A., Solinís, M. Á., Rodríguez-Gascón, A., Almeida, A. J., & Préat, V. Nanostructured lipid carriers: Promising drug delivery systems for future clinics. 12(1), 143-161. Nanomedicine: Nanotechnology, Biology and Medicine; 2016.
  29. Garud, A., Singh, D., & Garud, N. Solid lipid nanoparticles (SLN): Method, characterization, and applications. 1(11), 384-393. International Current Pharmaceutical Journal; 2012.
  30. Ekambaram, P., Sathali, A. H., & Priyanka, K. Solid lipid nanoparticles: A review. 2(1), 80-102. Scientific Reviews & Chemical Communications; 2012.
  31. Shah, R., Eldridge, D., Palombo, E., & Harding, I. H. Lipid nanoparticles: Production, characterization, and stability. 2(1-2), 47-58. Springer Science Reviews; 2014.
  32. Haider, M., Abdin, S. M., Kamal, L., & Orive, G. Nanostructured lipid carriers for delivery of chemotherapeutics: A review. 12(3), 288. Pharmaceutics; 2020.
  33. zur Mühlen, A., Schwarz, C., & Mehnert, W.  Solid lipid nanoparticles (SLN) for controlled drug delivery: Drug release and release mechanism. 45(2), 149-155. European Journal of Pharmaceutics and Biopharmaceutics; 1998.
  34. Kaur, P., Garg, T., Rath, G., & Goyal, A. K. Nanostructured lipid carriers: An emerging platform for improving oral bioavailability of poorly water-soluble drugs. 7, 43-51. Nanotechnology, Science, and Applications; 2014.
  35. Fang, R. H., Aryal, S., Hu, C. M. J., & Zhang, L. Quick synthesis of lipid-polymer hybrid nanoparticles with low polydispersity using a single-step sonication method. 29(30), 10408-10415. Langmuir; 2013.
  36. Sabale, P. M., Sabale, V. P., & Sabale, P. M. Enhancement of solubility and dissolution rate of poorly soluble anticancer drug. 58(4), 1184-1192. Indian Journal of Pharmaceutical Education and Research; 2024.
  37. Janakiraman, K., Madhav, K. V., Padavala, S. K. C., & Pamujula, N. H. Enhanced bioavailability through quality by design optimization of ceritinib nanostructured lipid carriers: Formulation, characterization, and stability evaluation. 14(11), 53-61. Journal of Applied Pharmaceutical Science; 2024.
  38.  Kumar, R., & Singh, A. Nanostructured lipid carriers: A novel approach for enhancing solubility and bioavailability of poorly soluble drugs. 7(5), 360-368. Journal of Emerging Technologies and Innovative Research; 2020.
  39. Patel, D., & Patel, M. Design, development, and characterization of nanostructured lipid carriers for enhancing the solubility and bioavailability of axitinib. 12(4), 45-52. Journal of Drug Delivery and Therapeutics; 2022.
  40. Jain, S., Jain, P., & Jain, A. K. Surface-engineered lipid-based nanoparticles as drug delivery systems for cancer therapeutics. 12(8), 1263-1279. Expert Opinion on Drug Delivery; 2015.
  41. Mishra, B., Patel, B. B., & Tiwari, S. Colloidal nanocarriers: A review on formulation technology, types and applications toward targeted drug delivery. 6(1), 9-24. Nanomedicine: Nanotechnology, Biology, and Medicine; 2010.
  42. Sengupta, S., & Eavarone, D. Targeted delivery of low-dose Doxil (PEGylated liposomal doxorubicin) using tumor-penetrating peptides. 10(1), 65-77. Cancer Cell; 2007.
  43. Lakhani, P., Patil, A., Chaudhuri, A., & Gupta, R. K.  Lipid-based drug delivery systems for delivery of poorly water-soluble drugs: Advances and challenges. 19(7), 2929-2942. AAPS PharmSciTech; 2018.
  44. Gottesman, M. M., Lavi, O., Hall, M. D., & Gillet, J. P. Toward a better understanding of the complexity of cancer drug resistance. 56, 85-102. Annual Review of Pharmacology and Toxicology; 2016
  45. Acharya, S., & Sahoo, S. K. PLGA nanoparticles containing various anticancer agents and their formulations. 149(3), 238-247. Journal of Controlled Release; 2011.
  46. Patel, N. R., & Patel, M. R. Targeting multidrug resistance in cancer with nanotechnology. 8(6), 2271-2281. International Journal of Pharmaceutical Sciences and Research; 2017.
  47. Sengupta, S., & Eavarone, D. Targeted delivery of low-dose Doxil (PEGylated liposomal doxorubicin) using tumor-penetrating peptides. 10(1), 65-77. Cancer Cell; 2007.
  48. Chauhan, A. S., Jain, R. K., & Pandey, H. Nanotechnology in cancer therapy: Role of lipid-based carriers in drug delivery. 9, 634-651. Journal of Cancer Therapy; 2018.
  49. Banerjee, R., & Sen, M. Lipid-based nanocarriers for drug delivery and bioavailability enhancement. 49(4), 304-311. Indian Journal of Biochemistry & Biophysics; 2012.
  50. Koudelka, Š., & Turánek, J. Liposomal paclitaxel formulations. 163(3), 322-334. Journal of Controlled Release; 2012.
  51. Rawal, S., Patel, M. M., & Maheshwari, R. Lipid-based nanocarriers in treatment of cancer: Emerging paradigms for overcoming multidrug resistance and enabling combination therapies. 11, 724579. Frontiers in Oncology; 2021.
  52. Mukherjee, S., Ray, S., & Thakur, R. S. Solid lipid nanoparticles: A modern formulation approach in drug delivery system. 71(4), 349-358. Indian Journal of Pharmaceutical Sciences; 2009.
  53. Peer, D., Karp, J. M., Hong, S., Farokhzad, O. C., Margalit, R., & Langer, R. Nanocarriers as an emerging platform for cancer therapy. 2(12), 751-760. Nature Nanotechnology; 2007.
  54. Muthu, M. S., Leong, D. T., Mei, L., & Feng, S. S. Nanotheranostics—application and further development of nanomedicine strategies for advanced theranostics. 4(6), 660-677. Theranostics; 2014.
  55. Gottesman, M. M., Lavi, O., Hall, M. D., & Gillet, J. P. Toward a better understanding of the complexity of cancer drug resistance. 56(1), 85–102. Annual Review of Pharmacology and Toxicology; 2016.
  56. Hanahan, D., & Weinberg, R. A. Hallmarks of cancer: The next generation. 144(5), 646–674. Cell; 2011.
  57. Longley, D. B., & Johnston, P. G. Molecular mechanisms of drug resistance. 205(2), 275–292. The Journal of Pathology; 2005.
  58. Zahreddine, H., & Borden, K. L. Mechanisms and insights into drug resistance in cancer. 4, 28. Frontiers in Pharmacology; 2013.
  59. Junttila, M. R., & de Sauvage, F. J. Influence of tumour micro-environment heterogeneity on therapeutic response. 501(7467), 346–354. Nature; 2013.
  60. Chen, Z., Shi, T., Zhang, L., et al. Mammalian drug efflux transporters of the ATP-binding cassette (ABC) family in multidrug resistance: 370(1), 153–164. A review of the past decade. Cancer Letters; 2016.
  61. Owais, M., Varshney, G. C., Choudhury, A., Chandra, S., & Gupta, C. M. Chloroquine encapsulated in malaria-infected erythrocyte-specific antibody-bearing liposomes effectively controls chloroquine-resistant Plasmodium berghei infections in mice. 39(1), 180–184. Antimicrobial Agents and Chemotherapy; 1995.
  62. Patel, N. R., & Patel, M. R. Targeting multidrug resistance in cancer with nanotechnology. 8(6), 2271–2281. International Journal of Pharmaceutical Sciences and Research; 2017.
  63. Patel, D., & Patel, M. Design, development, and characterization of nanostructured lipid carriers for enhancing the solubility and bioavailability of axitinib. 12(4), 45-52. Journal of Drug Delivery and Therapeutics; 2022.
  64. Choudhuri, S., & Klaassen, C. D. Structure, function, expression, genomic organization, and single nucleotide polymorphisms of human ABCB1 (MDR1): Progress in understanding multidrug resistance mediated by P-glycoprotein. 34(5), 695-701. Drug Metabolism and Disposition; 2006.
  65. Koudelka, Š., & Turánek, J. Liposomal paclitaxel formulations. 163(3), 322–334. Journal of Controlled Release; 2012.
  66. Rawal, S., Patel, M. M., & Maheshwari, R. Lipid-based nanocarriers in treatment of cancer: Emerging paradigms for overcoming multidrug resistance and enabling combination therapies. 11, 724579. Frontiers in Oncology; 2021.
  67. Jain, S., Jain, P., & Jain, A. K. Surface-engineered lipid-based nanoparticles as drug delivery systems for cancer therapeutics. 12(8), 1263-1279. Expert Opinion on Drug Delivery; 2015.
  68. Mukherjee, S., Ray, S., & Thakur, R. S. Solid lipid nanoparticles: A modern formulation approach in drug delivery system. 71(4), 349-358. Indian Journal of Pharmaceutical Sciences; 2009.
  69. Peer, D., Karp, J. M., Hong, S., Farokhzad, O. C., Margalit, R., & Langer, R. Nanocarriers as an emerging platform for cancer therapy. 2(12), 751-760. Nature Nanotechnology; 2007.
  70. Anjali Devi Nippani, Krishnaveni Janapareddi. Development and Ex-vivo Evaluation of Atorvastatin Microemlsions for Transdermal Delivery using box-Behnken Design. Volume 8(3). International Journal of Pharmacy and Biological Sciences; 2018.
  71. Muthu, M. S., Leong, D. T., Mei, L., & Feng, S. S. Nanotheranostics—application and further development of nanomedicine strategies for advanced theranostics. 4(6), 660-677. Theranostics; 2014.

Reference

  1. Gottesman, M. M., Lavi, O., Hall, M. D., & Gillet, J. P. Toward a better understanding of the complexity of cancer drug resistance. 56(1), 85–102. Annual Review of Pharmacology and Toxicology; 2016.
  2. Hanahan, D., & Weinberg, R. A. Hallmarks of cancer: The next generation. 144(5), 646–674. Cell; 2011.
  3. Junttila, M. R., & de Sauvage, F. J. Influence of tumour micro-environment heterogeneity on therapeutic response. 501(7467), 346–354. Nature; 2013.
  4. Zahreddine, H., & Borden, K. L. Mechanisms and insights into drug resistance in cancer. 4, 28. Frontiers in Pharmacology; 2013.
  5. Longley, D. B., & Johnston, P. G. Molecular mechanisms of drug resistance. 205(2), 275–292. The Journal of Pathology; 2005.
  6. Owais, M., Varshney, G. C., Choudhury, A., Chandra, S., & Gupta, C. M. Chloroquine encapsulated in malaria-infected erythrocyte-specific antibody-bearing liposomes effectively controls chloroquine-resistant Plasmodium berghei infections in mice. 39(1), 180–184. Antimicrobial Agents and Chemotherapy; 1995.
  7. Ansari, M. T., Ramlan, T. A., Jamaluddin, N. N., Zamri, N., Salfi, R., Khan, A., Sami, F., Majeed, S., & Hasnain, M. S. Lipid-based nanocarriers for cancer and tumor treatment. 26(34), 4272–4276. Current Pharmaceutical Design; 2020.
  8. Anwar, D. M., Hedeya, H. Y., Ghozlan, S. H., Ewas, B. M., & Khattab, S. N. Surface-modified lipid-based nanocarriers as a pivotal delivery approach for cancer therapy: Application and recent advances in targeted cancer treatment. 13, Article number: 106. Beni-Suef University Journal of Basic and Applied Sciences; 2024.
  9. Dechbumroong, P., Hu, R., & Liang, X.-J. Recent advanced lipid-based nanomedicines for overcoming cancer resistance. 7, 24. Cancer Drug Resistance; 2024.
  10. Benko, A., Medina-Cruz, D., & Webster, T. J. Nanocarrier drug resistant tumor interactions: Novel approaches to fight drug resistance in cancer. 4, 264–297. Cancer Drug Resistance; 2021.
  11. Chu, S., & Stochaj, U. Exploring near-infrared absorbing nanocarriers to overcome cancer drug resistance. 3, 302–333. Cancer Drug Resistance; 2020.
  12. Gottesman, M. M., & Pastan, I. Biochemistry of multidrug resistance mediated by the multidrug transporter. 62(1), 385-427. Annual Review of Biochemistry; 1993.
  13. Aller, S. G., Yu, J., Ward, A., et al. Structure of P-glycoprotein reveals a molecular basis for polyspecific drug binding. 323(5922), 1718-1722. Science; 2009.
  14. Fletcher, J. I., Haber, M., Henderson, M. J., & Norris, M. D. ABC transporters in cancer: More than just drug efflux pumps. 10(2), 147-156. Nature Reviews Cancer; 2010.
  15. Chen, Z., Shi, T., Zhang, L., et al. Mammalian drug efflux transporters of the ATP-binding cassette (ABC) family in multidrug resistance: A review of the past decade. 370(1), 153-164. Cancer Letters; 2016.
  16. Lin, J. H., & Yamazaki, M. Role of P-glycoprotein in pharmacokinetics: Clinical implications. 42(1), 59-98. Clinical Pharmacokinetics; 2003.
  17. Choudhuri, S., & Klaassen, C. D. Structure, function, expression, genomic organization, and single nucleotide polymorphisms of human ABCB1 (MDR1): Progress in understanding multidrug resistance mediated by P-glycoprotein. 34(5), 695-701. Drug Metabolism and Disposition; 2006.
  18. Seelig, A.  A general pattern for substrate recognition by P-glycoprotein. 251(1-2), 252-261. European Journal of Biochemistry; 1998.
  19. Choudhury, A., Sonowal, K., Laskar, R. E., Deka, D., & Dey, B. K. Liposome: A carrier for effective drug delivery. 10(3), 115-122. Journal of Advanced Pharmaceutical Technology & Research; 2019.
  20. Anjima, K. K., Akshaya, P., Ramalingam, N., & Vimal, K. R. Liposomal gene delivery system. 10(2), 25-32. International Journal of Pharmaceutical and Phytopharmacological Research; 2020.
  21. Agarwal, K. Liposome-assisted drug delivery: An updated review. 84(4), 322-334. Indian Journal of Pharmaceutical Sciences; 2022.
  22. Kulkarni, P. R., Yadav, J. D., & Vaidya, K. A. Liposomes: A novel drug delivery system. 3(2), 45-52. International Journal of Current Pharmaceutical Review and Research; 2011.
  23. Dhone, P. Liposomes, protein, and peptide drug delivery systems. 15(1), 12-20. International Journal of Pharmaceutical Sciences; 2024.
  24. Pawar, V., Mishra, K. S., Yadav, M., & Tiwari, A. Liposome: A powerful approach festinates drug delivery system. 3(1), 14-21. International Journal of Drug Delivery Technology; 2013.
  25. Kshirsagar, N. A., Pandya, S. K., Kirodian, B. G., & Sanath, S. Liposomal drug delivery system from laboratory to clinic. 51(3), 209-215. Journal of Postgraduate Medicine; 2005.
  26. Mehnert, W., & Mäder, K. Solid lipid nanoparticles: Production, characterization, and applications. 47(2-3), 165-196. Advanced Drug Delivery Reviews; 2001.
  27. Müller, R. H., Radtke, M., & Wissing, S. A. Solid lipid nanoparticles (SLN) and nanostructured lipid carriers (NLC) in cosmetic and dermatological preparations. 54(Suppl 1), S131-S155. Advanced Drug Delivery Reviews; 2002.
  28. Beloqui, A., Solinís, M. Á., Rodríguez-Gascón, A., Almeida, A. J., & Préat, V. Nanostructured lipid carriers: Promising drug delivery systems for future clinics. 12(1), 143-161. Nanomedicine: Nanotechnology, Biology and Medicine; 2016.
  29. Garud, A., Singh, D., & Garud, N. Solid lipid nanoparticles (SLN): Method, characterization, and applications. 1(11), 384-393. International Current Pharmaceutical Journal; 2012.
  30. Ekambaram, P., Sathali, A. H., & Priyanka, K. Solid lipid nanoparticles: A review. 2(1), 80-102. Scientific Reviews & Chemical Communications; 2012.
  31. Shah, R., Eldridge, D., Palombo, E., & Harding, I. H. Lipid nanoparticles: Production, characterization, and stability. 2(1-2), 47-58. Springer Science Reviews; 2014.
  32. Haider, M., Abdin, S. M., Kamal, L., & Orive, G. Nanostructured lipid carriers for delivery of chemotherapeutics: A review. 12(3), 288. Pharmaceutics; 2020.
  33. zur Mühlen, A., Schwarz, C., & Mehnert, W.  Solid lipid nanoparticles (SLN) for controlled drug delivery: Drug release and release mechanism. 45(2), 149-155. European Journal of Pharmaceutics and Biopharmaceutics; 1998.
  34. Kaur, P., Garg, T., Rath, G., & Goyal, A. K. Nanostructured lipid carriers: An emerging platform for improving oral bioavailability of poorly water-soluble drugs. 7, 43-51. Nanotechnology, Science, and Applications; 2014.
  35. Fang, R. H., Aryal, S., Hu, C. M. J., & Zhang, L. Quick synthesis of lipid-polymer hybrid nanoparticles with low polydispersity using a single-step sonication method. 29(30), 10408-10415. Langmuir; 2013.
  36. Sabale, P. M., Sabale, V. P., & Sabale, P. M. Enhancement of solubility and dissolution rate of poorly soluble anticancer drug. 58(4), 1184-1192. Indian Journal of Pharmaceutical Education and Research; 2024.
  37. Janakiraman, K., Madhav, K. V., Padavala, S. K. C., & Pamujula, N. H. Enhanced bioavailability through quality by design optimization of ceritinib nanostructured lipid carriers: Formulation, characterization, and stability evaluation. 14(11), 53-61. Journal of Applied Pharmaceutical Science; 2024.
  38.  Kumar, R., & Singh, A. Nanostructured lipid carriers: A novel approach for enhancing solubility and bioavailability of poorly soluble drugs. 7(5), 360-368. Journal of Emerging Technologies and Innovative Research; 2020.
  39. Patel, D., & Patel, M. Design, development, and characterization of nanostructured lipid carriers for enhancing the solubility and bioavailability of axitinib. 12(4), 45-52. Journal of Drug Delivery and Therapeutics; 2022.
  40. Jain, S., Jain, P., & Jain, A. K. Surface-engineered lipid-based nanoparticles as drug delivery systems for cancer therapeutics. 12(8), 1263-1279. Expert Opinion on Drug Delivery; 2015.
  41. Mishra, B., Patel, B. B., & Tiwari, S. Colloidal nanocarriers: A review on formulation technology, types and applications toward targeted drug delivery. 6(1), 9-24. Nanomedicine: Nanotechnology, Biology, and Medicine; 2010.
  42. Sengupta, S., & Eavarone, D. Targeted delivery of low-dose Doxil (PEGylated liposomal doxorubicin) using tumor-penetrating peptides. 10(1), 65-77. Cancer Cell; 2007.
  43. Lakhani, P., Patil, A., Chaudhuri, A., & Gupta, R. K.  Lipid-based drug delivery systems for delivery of poorly water-soluble drugs: Advances and challenges. 19(7), 2929-2942. AAPS PharmSciTech; 2018.
  44. Gottesman, M. M., Lavi, O., Hall, M. D., & Gillet, J. P. Toward a better understanding of the complexity of cancer drug resistance. 56, 85-102. Annual Review of Pharmacology and Toxicology; 2016
  45. Acharya, S., & Sahoo, S. K. PLGA nanoparticles containing various anticancer agents and their formulations. 149(3), 238-247. Journal of Controlled Release; 2011.
  46. Patel, N. R., & Patel, M. R. Targeting multidrug resistance in cancer with nanotechnology. 8(6), 2271-2281. International Journal of Pharmaceutical Sciences and Research; 2017.
  47. Sengupta, S., & Eavarone, D. Targeted delivery of low-dose Doxil (PEGylated liposomal doxorubicin) using tumor-penetrating peptides. 10(1), 65-77. Cancer Cell; 2007.
  48. Chauhan, A. S., Jain, R. K., & Pandey, H. Nanotechnology in cancer therapy: Role of lipid-based carriers in drug delivery. 9, 634-651. Journal of Cancer Therapy; 2018.
  49. Banerjee, R., & Sen, M. Lipid-based nanocarriers for drug delivery and bioavailability enhancement. 49(4), 304-311. Indian Journal of Biochemistry & Biophysics; 2012.
  50. Koudelka, Š., & Turánek, J. Liposomal paclitaxel formulations. 163(3), 322-334. Journal of Controlled Release; 2012.
  51. Rawal, S., Patel, M. M., & Maheshwari, R. Lipid-based nanocarriers in treatment of cancer: Emerging paradigms for overcoming multidrug resistance and enabling combination therapies. 11, 724579. Frontiers in Oncology; 2021.
  52. Mukherjee, S., Ray, S., & Thakur, R. S. Solid lipid nanoparticles: A modern formulation approach in drug delivery system. 71(4), 349-358. Indian Journal of Pharmaceutical Sciences; 2009.
  53. Peer, D., Karp, J. M., Hong, S., Farokhzad, O. C., Margalit, R., & Langer, R. Nanocarriers as an emerging platform for cancer therapy. 2(12), 751-760. Nature Nanotechnology; 2007.
  54. Muthu, M. S., Leong, D. T., Mei, L., & Feng, S. S. Nanotheranostics—application and further development of nanomedicine strategies for advanced theranostics. 4(6), 660-677. Theranostics; 2014.
  55. Gottesman, M. M., Lavi, O., Hall, M. D., & Gillet, J. P. Toward a better understanding of the complexity of cancer drug resistance. 56(1), 85–102. Annual Review of Pharmacology and Toxicology; 2016.
  56. Hanahan, D., & Weinberg, R. A. Hallmarks of cancer: The next generation. 144(5), 646–674. Cell; 2011.
  57. Longley, D. B., & Johnston, P. G. Molecular mechanisms of drug resistance. 205(2), 275–292. The Journal of Pathology; 2005.
  58. Zahreddine, H., & Borden, K. L. Mechanisms and insights into drug resistance in cancer. 4, 28. Frontiers in Pharmacology; 2013.
  59. Junttila, M. R., & de Sauvage, F. J. Influence of tumour micro-environment heterogeneity on therapeutic response. 501(7467), 346–354. Nature; 2013.
  60. Chen, Z., Shi, T., Zhang, L., et al. Mammalian drug efflux transporters of the ATP-binding cassette (ABC) family in multidrug resistance: 370(1), 153–164. A review of the past decade. Cancer Letters; 2016.
  61. Owais, M., Varshney, G. C., Choudhury, A., Chandra, S., & Gupta, C. M. Chloroquine encapsulated in malaria-infected erythrocyte-specific antibody-bearing liposomes effectively controls chloroquine-resistant Plasmodium berghei infections in mice. 39(1), 180–184. Antimicrobial Agents and Chemotherapy; 1995.
  62. Patel, N. R., & Patel, M. R. Targeting multidrug resistance in cancer with nanotechnology. 8(6), 2271–2281. International Journal of Pharmaceutical Sciences and Research; 2017.
  63. Patel, D., & Patel, M. Design, development, and characterization of nanostructured lipid carriers for enhancing the solubility and bioavailability of axitinib. 12(4), 45-52. Journal of Drug Delivery and Therapeutics; 2022.
  64. Choudhuri, S., & Klaassen, C. D. Structure, function, expression, genomic organization, and single nucleotide polymorphisms of human ABCB1 (MDR1): Progress in understanding multidrug resistance mediated by P-glycoprotein. 34(5), 695-701. Drug Metabolism and Disposition; 2006.
  65. Koudelka, Š., & Turánek, J. Liposomal paclitaxel formulations. 163(3), 322–334. Journal of Controlled Release; 2012.
  66. Rawal, S., Patel, M. M., & Maheshwari, R. Lipid-based nanocarriers in treatment of cancer: Emerging paradigms for overcoming multidrug resistance and enabling combination therapies. 11, 724579. Frontiers in Oncology; 2021.
  67. Jain, S., Jain, P., & Jain, A. K. Surface-engineered lipid-based nanoparticles as drug delivery systems for cancer therapeutics. 12(8), 1263-1279. Expert Opinion on Drug Delivery; 2015.
  68. Mukherjee, S., Ray, S., & Thakur, R. S. Solid lipid nanoparticles: A modern formulation approach in drug delivery system. 71(4), 349-358. Indian Journal of Pharmaceutical Sciences; 2009.
  69. Peer, D., Karp, J. M., Hong, S., Farokhzad, O. C., Margalit, R., & Langer, R. Nanocarriers as an emerging platform for cancer therapy. 2(12), 751-760. Nature Nanotechnology; 2007.
  70. Anjali Devi Nippani, Krishnaveni Janapareddi. Development and Ex-vivo Evaluation of Atorvastatin Microemlsions for Transdermal Delivery using box-Behnken Design. Volume 8(3). International Journal of Pharmacy and Biological Sciences; 2018.
  71. Muthu, M. S., Leong, D. T., Mei, L., & Feng, S. S. Nanotheranostics—application and further development of nanomedicine strategies for advanced theranostics. 4(6), 660-677. Theranostics; 2014.

Photo
P. Sushma
Corresponding author

MB School of Pharmaceutical Sciences, Mohan Babu University

Photo
S. Varalaxmi
Co-author

MB School of Pharmaceutical Sciences, Mohan Babu University

P. Sushma*, S. Varalaxmi, Role Of Lipid Carriers in Overcoming Drug Resistance in Cancer, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 6, 5294-5308. https://doi.org/10.5281/zenodo.15755721

More related articles
Formulation, Evaluation, and Applications of Omepr...
Amit Adhikari, Dr. Amit Chaudhary, Anas Khan, ...
An In Depth Exploration of Ginkgo Biloba: A Review...
Shreya Talreja, Dr Shashank Tiwari, ...
A Review of Parkinson Disease...
Payal Sahu, Mayur Hirve, Anshika Singh Chauhan, Enti Srivalli, Su...
Pharmacognosy Of Essntial Oils: Recent Advances in Extraction and Therapeutic Ap...
Rahul Dubey, Vijendra Baghel, Samriddhi Gidion, Vasant Harjpal, ...
Artificial Intelligence: A Review on AI Powered Applications in Pharmacy ...
Dr. Shital Dange, Vaishnavi Latthe, Samruddhi Paramane, Sanika Patil, Arpita Mali, Triveni Rugge, ...
Preliminary Phytochemical Screening, Photoluminance Study And TLC Of Leaf Extrac...
Nikhil Rana, Chinu Kumari, Dev Prakash Dahiya, Rahul Sharma, ...
Related Articles
A Review on Liposomal Insitu Gel Formulations for Intranasal Delivery of Antidep...
Gagan K. S., S. Swetha Malika Devi, Beny Baby, S Rajarajan, Yallappa, ...
Formulation And Evaluation of Hibiscus Tea Granules Powder...
Jyoti Mandal, Anil Sori, Tulsidas Nimbekar, ...
A Review on Orally Administration of Lipospheres for Enhancement of Solubility...
Neeraj Kumar Rathour, Ajay Pratap Singh, Saurabh Mishra, ...
Formulation And Evaluation Of Antiperspirant/Hyperhidrosis Solution...
Ang Solanki, Nripen Prakash Khare , Khushbu Pinjane, Satyaendra Shrivastava , ...
More related articles
An In Depth Exploration of Ginkgo Biloba: A Review...
Shreya Talreja, Dr Shashank Tiwari, ...
A Review of Parkinson Disease...
Payal Sahu, Mayur Hirve, Anshika Singh Chauhan, Enti Srivalli, Suchita Wamankar, Dr. Gyanesh Kumar S...
An In Depth Exploration of Ginkgo Biloba: A Review...
Shreya Talreja, Dr Shashank Tiwari, ...
A Review of Parkinson Disease...
Payal Sahu, Mayur Hirve, Anshika Singh Chauhan, Enti Srivalli, Suchita Wamankar, Dr. Gyanesh Kumar S...