scispace - formally typeset
Search or ask a question

What are the disadvantages of liposomal doxorubicin? 


Best insight from top research papers

Liposomal doxorubicin, while offering advantages like reduced cardiotoxicity and improved tumor targeting, has some drawbacks. One limitation is the potential for fast clearance, necessitating further investigation into optimizing factors like ligand density, stability, and release rate . Additionally, specific modifications, such as sialic acid (SA) on liposomes, can impact drug retention properties and, consequently, the antitumor effect . Moreover, the intracellular uptake of doxorubicin from liposomal formulations is lower compared to free doxorubicin, potentially affecting its overall efficacy in certain cancer cell lines . These factors highlight the need for ongoing research to enhance the efficiency and effectiveness of liposomal doxorubicin delivery systems.

Answers from top 5 papers

More filters
Papers (5)Insight
Liposomal doxorubicin has reduced cardiotoxicity compared to regular doxorubicin, but disadvantages are not specified in the provided data.
Liposomal doxorubicin (Doxil®) disadvantages include cardiotoxicity and skin accumulation. Targeting with TIMP3 peptide enhances tumour cell uptake, potentially reducing side effects.
Open accessJournal ArticleDOI
Kenan Aloss, Péter Hamar 
01 Mar 2023-Pharmaceutics
3 Citations
Liposomal doxorubicin's disadvantages include limited efficacy compared to conventional doxorubicin, requiring further investigations on fast clearance, ligand density optimization, stability, and release rate for enhanced delivery.
Liposomal doxorubicin can have dose-limiting toxicities, but modifications like sialic acid-cholesterol conjugates can enhance tumor targeting and reduce side effects, improving efficacy.
Liposomal doxorubicin has lower intracellular uptake compared to free doxorubicin, limiting its effectiveness in treating hepatocellular carcinoma due to inadequate exposure levels in resistant cell lines.

Related Questions

What is various doxorubicine ?3 answersDoxorubicin is an antitumor drug that is widely used in the treatment of various human cancers. It has a broad spectrum of antitumor activity and is considered one of the most utilized antitumor drugs worldwide. Efforts have been made to develop analogs of doxorubicin that have less cardiotoxicity and greater antitumor efficacy. Several analogs, such as epirubicin, esorubicin, idarubicin, and menogaril, have been studied extensively and show promise in terms of reduced cardiotoxicity and increased antitumor activity. Additionally, research has focused on combining doxorubicin with other compounds to enhance its effectiveness. For example, a polypeptide fragment of the epidermal growth factor has been conjugated with doxorubicin to selectively target cancer cells and reduce resistance to the drug. Overall, doxorubicin and its analogs continue to be an area of active research in the field of cancer treatment.
What are the advantages and disadvantages of using extracellular vesicles as a drug delivery system?5 answersExtracellular vesicles (EVs) have several advantages as a drug delivery system. They have characteristics similar to parent cells, making them suitable for intercellular communication and modulation of disease microenvironment. EVs can cross extracellular and intracellular barriers, making them superior to synthetic nanoparticles. They also have the ability to transport beneficial biomolecules among distant cells. EVs are low in toxicity, low in immunogenicity, and have high stability, delivery efficiency, and biocompatibility. They can also target specific sites in the brain due to their ability to cross the blood-brain barrier. EVs can protect and deliver a functional cargo using endogenous cellular mechanisms, making them attractive as therapeutics. However, there are also some challenges associated with using EVs as a drug delivery system. Developing a consistent biochemical strategy for their clinical therapeutic use can be difficult. Additionally, there are barriers associated with brain-targeted drug delivery using EVs. Further research is needed to fully understand the mechanisms behind the therapeutic benefits of EVs.
What are the advantages and disadvantages of each method of liposome preparation?5 answersLiposome preparation methods have both advantages and disadvantages. Conventional methods are easy to implement and do not require complex equipment, but they have low efficiency in encapsulating active pharmaceutical ingredients (APIs) within liposomes and scaling up technological processes is complex. Novel methods, such as microfluidic techniques, offer highly controlled processes, high reproducibility, and scalability to industrial production, making them suitable for encapsulating different APIs. However, these methods may require more sophisticated equipment and expertise. Both conventional and novel methods have their benefits and limitations, and the choice of method depends on the specific requirements of liposome production.
What are the challenges associated with the use of tumor-derived extracellular vesicles?5 answersTumor-derived extracellular vesicles (EVPs) pose several challenges in their use. Firstly, EVPs have been found to induce fatty liver and enhance toxicities of chemotherapy. This can have negative effects on the overall health of the patient and may limit the effectiveness of treatment. Secondly, the cargo content of EVPs from tumor cells can be dysregulated in certain conditions, such as obesity, leading to elevated amounts of pro-inflammatory proteins, fatty acids, and carcinogenic microRNAs. This dysregulation can contribute to tumor progression and may complicate the use of EVPs as biomarkers or therapeutic agents. Lastly, there are limitations and challenges in EV-based research, including the need for further understanding of their biogenesis, physical features, and cargo. These challenges must be addressed in order to fully harness the potential of tumor-derived EVPs in clinical applications.
What are the cytotoxic effects of doxorubicin in cell viability?1 answersDoxorubicin has cytotoxic effects on cell viability. It induces DNA damage, inhibits cell proliferation, impairs mitochondria, and leads to cell death. The cardiotoxicity of doxorubicin is attributed to the production of superoxide anion inside the cell. Cells with reduced superoxide dismutase (SOD) activity are more susceptible to doxorubicin-induced cytotoxicity. Neutrophils produce superoxide in response to doxorubicin, leading to cytotoxic effects. In feline cancer cells, doxorubicin combined with salinomycin decreases cell viability, with a synergistic effect observed at achievable doxorubicin concentrations. Aptamers targeting EpCAM, a cancer biomarker, can deliver doxorubicin to epithelial ovarian cancer cells, resulting in cytotoxicity.
How to load doxorubicin in liposomes?3 answersDoxorubicin can be loaded into liposomes using different methods. One method is remote loading, where doxorubicin is encapsulated in liposomes using an ammonium sulfate gradient method. Another method is co-encapsulation, where doxorubicin is co-encapsulated with other drugs in liposomes. Additionally, liposomes can be drawn into nanotubes, forming a unique structure called DoxL-PAuNT, which allows for efficient release of doxorubicin. Another approach is to load doxorubicin-carrying liposomes onto the surfaces of macrophages, utilizing their active tumor-targeting capability. These different methods provide options for loading doxorubicin into liposomes, allowing for controlled and targeted delivery of the drug to tumor sites.

See what other people are reading

How have previous studies explored the role of astrocytes in Aβ clearance through autophagy and phagocytosis?
5 answers
Previous studies have extensively investigated the role of astrocytes in Aβ clearance through autophagy and phagocytosis in Alzheimer's disease (AD). Astrocytes have been found to contribute significantly to Aβ clearance and degradation. Specifically, astrocytes play a crucial role in engulfing and degrading Aβ aggregates, preventing their accumulation and subsequent neurotoxic effects. Additionally, astrocytes have been shown to aid in the clearance of amyloid plaques and support synaptic plasticity, highlighting their protective role in AD pathology. Furthermore, studies have identified specific mechanisms, such as the involvement of LRP4 in promoting Aβ uptake by astrocytes, emphasizing the importance of astrocytic function in Aβ pathology and cognitive function. These findings underscore the potential therapeutic implications of targeting astrocytes to enhance Aβ clearance mechanisms in AD.
Who cited the meta-analysis done by Abreu-Ledón et al. (2018)?
5 answers
The meta-analysis conducted by Abreu-Ledón et al. (2018) was cited by Ronney Abaza in his study on the responsible application of minimally invasive surgery to complex surgeries, particularly focusing on robotic management of inferior vena cava thrombi. Abaza highlighted the importance of gradually progressing from less difficult to more challenging procedures with strict patient selection, drawing on the successful application of minimally invasive techniques in such complex surgeries. By referencing the meta-analysis, Abaza emphasizes the significance of evidence-based approaches in advancing surgical innovations and ensuring patient safety in high-risk procedures.
What are the mechanisms by which gold compounds exhibit antitumor activity?
5 answers
Gold compounds exhibit antitumor activity through various mechanisms. They primarily target thioredoxin reductase (TrxR), triggering cell death via reactive oxygen species (ROS). Additionally, gold complexes induce DNA damage, autophagy, and apoptosis in cancer cells. Some gold compounds have been shown to inhibit cancer and endothelial cell migration, as well as angiogenesis. Moreover, gold compounds have been reported to reverse tumor immune escape and enhance immune cell functions, contributing to enhanced anticancer effects. Overall, the multifaceted mechanisms of action of gold compounds make them promising candidates for cancer therapy, with potential for further development and clinical application.
What is the level of TFEB in psoriasis?
5 answers
The level of Transcription Factor EB (TFEB) in psoriasis is not directly addressed in the provided contexts. However, TFEB is a master regulator of autophagy and lysosomal biogenesis, with potential implications in various biological processes. In psoriasis, studies have focused on factors like Vascular Endothelial Growth Factor (VEGF), Transforming Growth Factor β1 (TGF-β1), and Pentraxin 3 (PTX 3) to understand disease severity and pathogenesis. These studies suggest that TFEB, although not directly measured, could play a role in the complex pathophysiology of psoriasis. Further research specifically investigating TFEB levels in psoriasis patients could provide valuable insights into its involvement in the disease process.
What are the administration routes of chemotherapeutics with nanocarriers in the treatment of colorectal cancer?
7 answers
In the treatment of colorectal cancer (CRC), nanocarriers have revolutionized the administration routes of chemotherapeutics, offering targeted and efficient delivery methods that aim to minimize side effects and enhance therapeutic efficacy. The primary administration routes for these nanocarrier-based chemotherapeutics include oral administration and targeted delivery systems, each with its unique mechanisms and advantages. Oral administration is highlighted as an attractive approach for CRC therapy due to its potential to improve the efficacy of local drug delivery while reducing systemic toxicity. This route faces challenges such as poor drug solubility, stability, and permeability, but nanotechnology offers solutions to overcome these barriers. Chitosan-based nanocarriers, for example, have been developed to enhance stability, targeting, and bioavailability of anti-CRC agents, providing a promising strategy for oral drug delivery. The unique physicochemical properties of nanoparticles assist in overcoming oral delivery challenges, including protection against gastrointestinal degradation. Targeted delivery systems, on the other hand, utilize nanocarriers functionalized with monoclonal antibodies or other targeting agents to actively direct chemotherapeutics to CRC cells. This approach leverages the enhanced permeability and retention (EPR) effect and specific molecular markers over-expressed on CRC cells to increase tumor accumulation and reduce adverse effects on healthy tissues. Surface-engineered nanocarriers can facilitate receptor-mediated deliveries, targeting overexpressed receptors in the CRC microenvironment for precise drug delivery. Additionally, magneto-fluorescent nanoparticles modified with targeting agents have shown potential for targeted nanotherapy, indicating the versatility of nanocarriers in delivering drugs directly to tumor sites. These administration routes, oral and targeted delivery, represent the forefront of nanocarrier-based chemotherapeutic delivery in CRC treatment. They exemplify the ongoing efforts to enhance the specificity, efficacy, and safety of cancer therapies through advanced nanotechnology.
How does Autotoxicity Work for seagrasses in marine organisms?
5 answers
Autotoxicity in seagrasses involves the negative impact of high ammonium concentrations on these marine organisms. Seagrasses like Zostera species face toxic effects due to ammonium enrichment, which can lead to internal accumulation of ammonium, affecting processes like energy metabolism, carbon fixation, and photosynthesis. The interaction between ammonium and other environmental stressors intensifies the toxic effects, with factors like light reduction and hyposaline stress exacerbating the detrimental outcomes of high ammonium levels on seagrasses. This highlights the importance of understanding how multiple stressors interact to predict and manage the impact of pollutants like ammonium on seagrass ecosystems in the face of increasing coastal pollution.
Doxorubicin liposomes with colorectal cancer
5 answers
Doxorubicin-loaded liposomes have shown promising results in the treatment of colorectal cancer. These liposomes can be engineered with specific peptides to enhance tumor-targeted drug delivery. Additionally, combining doxorubicin-loaded liposomes with nanoparticle-based photothermal therapy (PTT) has demonstrated improved outcomes in mouse models of colorectal cancer. The use of biocompatible lipopolyplexes, which encapsulate polyplexes containing FAK siRNA and doxorubicin, has also been explored as a potential treatment strategy for colorectal cancer. Overall, these studies highlight the efficacy of liposomal doxorubicin in targeting colorectal cancer cells and the potential of combining it with innovative therapies to enhance treatment outcomes.
What is the effective concentraion of carbon dots in cells for drug delivery?
5 answers
The effective concentration of carbon dots (CDs) in cells for drug delivery varies based on the specific application. CDs have shown potential as drug delivery vehicles due to their biocompatibility, small size, and unique physicochemical properties. Studies have demonstrated that CDs can enhance drug solubility, bioavailability, and cellular uptake. Additionally, CDs have been found to selectively enhance cell internalization of drugs like doxorubicin, increasing their cytotoxicity and acting as drug sensitizers. However, it is crucial to consider factors like biosafety, toxicity, and immune system impact when determining the optimal concentration of CDs for drug delivery. Further research is needed to establish the ideal concentration of CDs in cells for different therapeutic applications, such as anti-inflammatory, antibacterial, and anticancer drug delivery.
How do nanocarriers affect the delivery and efficacy of chemo-radiation therapy in treating colorectal cancer?
10 answers
Nanocarriers have significantly impacted the delivery and efficacy of chemo-radiation therapy in treating colorectal cancer (CRC) by enhancing the precision, reducing side effects, and improving patient outcomes. These nanoscale delivery systems are engineered to improve the stability, selectivity, and efficacy of anti-colon cancer drugs, thereby pushing the boundaries of CRC therapy to new heights. They offer a promising alternative to conventional treatments, which are often associated with aggressive side effects and a lack of target specificity, by enabling targeted therapy that can distinguish between healthy and cancerous tissues, thus potentially reducing patient side effects. Nanocarriers, such as polymeric nanoparticles, liposomes, and solid lipid nanoparticles, have been developed to directly target cancer cells and regulate drug release. This targeted approach not only enhances therapeutic efficacy but also minimizes side effects by ensuring that the cytotoxic drugs accumulate more efficiently in the tumor cells rather than in healthy tissues. The versatility of nanocarriers allows for the co-delivery of chemotherapeutic drugs and photosensitizers, as demonstrated in studies where lipid-polymer hybrid nanoparticles were used to combine X-ray induced photodynamic therapy with chemotherapy, showing enhanced anti-tumor effects in CRC cells. Furthermore, the surface engineering of nanocarriers enables receptor-mediated delivery to overexpressed receptors in the CRC microenvironment, enhancing the specificity and effectiveness of the treatment. Innovative approaches, such as the development of nanoparticles that can improve tumor cytotoxicity and reduce systemic toxicity, have shown promising results in both in vitro and in vivo models, suggesting a potential for novel targeted delivery systems that could significantly improve the outcomes of CRC therapy. Recent research has also explored the synergistic effects of nanocarriers with anticancer drugs, highlighting the potential for nanomaterials to activate the immune microenvironment and enhance the efficacy of treatments through improved drug bioavailability and targeted delivery. These advancements in nanotechnology and the successful application of nanocarriers in clinical trials underscore their critical role in revolutionizing the treatment landscape for colorectal cancer.
What is iodide intracellular concentration of human cells?
4 answers
The intracellular iodide concentration in human cells varies depending on the cell type and conditions. Studies have shown that normal human thyroid cells can concentrate iodide even when dispersed, indicating that intact thyroid follicles are not essential for this process. In contrast, the uptake of 6-125I-iodo-2-methyl-1,4-naphthoquinol bis(diammonium phosphate) in malignant cells was significantly higher than in normal cells, with uptakes ranging from 0.89 to 11.3 pCi/cell. Additionally, intrathyroidal iodine concentrations were found to change after the administration of non-ionic X-ray contrast media, with the potential for iodine accumulation in various tissues, including the liver, brain, kidney, and skin. These findings highlight the dynamic nature of iodide concentrations within human cells under different physiological and pathological conditions.
What is the current understanding of the physiological effects of piretrine and pyrethroid exposure on humans?
5 answers
Current research indicates that exposure to pyrethroids like allethrin and prallethrin can induce various physiological effects in humans. Pyrethroids, synthetic derivatives of natural pyrethrins, primarily target the sodium channels in insect neurons, potentially leading to toxicity in vertebrates, including humans. Acute exposure to pyrethroids can enhance the late sodium current in the heart, causing cardiac arrhythmias, while long-term exposure may result in oxidative stress and tissue remodeling. Furthermore, studies have shown that exposure to pyrethroids equivalent to human consumption levels can lead to systemic and reproductive toxicity, impacting organ function, antioxidant status, and anatomical structures. These findings emphasize the importance of understanding the potential health risks associated with pyrethroid exposure in humans.