scispace - formally typeset
Search or ask a question

Cancer therapies/therapeutics using nanoparticles/nanomedicine and biomaterials targeting the tumor microenvironment for cancer treatment 


Best insight from top research papers

Nanoparticles carrying chemotherapeutic drugs have shown promise in reducing side effects of cancer treatments . Additionally, targeting the tumor microenvironment is crucial for effective cancer therapy. Research has highlighted the significance of modulating immune responses within the tumor microenvironment for successful treatment. Strategies such as blocking TGF-β signaling in CD4+ T cells have been found to remodel the tumor microenvironment and inhibit cancer progression . Moreover, inhibiting lipid synthesis in regulatory T cells (Treg cells) through SREBP signaling pathways has shown to enhance antitumor immune responses without autoimmune toxicity . Furthermore, disrupting the premetastatic microenvironment using low-dose adjuvant epigenetic therapy has been effective in inhibiting the formation and growth of metastases post-surgery . These approaches demonstrate the potential of nanomedicine and biomaterials in targeting the tumor microenvironment for improved cancer treatment outcomes.

Answers from top 5 papers

More filters
Papers (5)Insight
Not addressed in the paper.
Not addressed in the paper.
Journal ArticleDOI
17 May 2021-Nature
4 Citations
Nanoparticles carrying chemotherapeutic drugs offer potential for targeted cancer treatment, aiming to reduce side effects by delivering drugs directly to tumors within the microenvironment.
Not addressed in the paper.
Not addressed in the paper.

Related Questions

How is tumor cell targeted by nanoparticle for drug delivery?5 answersNanoparticles are utilized for targeted drug delivery to tumor cells by decorating them with functional ligands that bind to specific receptors overexpressed on cancer cell surfaces. These ligands include transferrin, folic acid, polypeptides, and hyaluronic acid, enhancing the specificity of drug delivery systems. Additionally, nanosystems with cancer-targeted ligands exploit cell surface-specific receptors, tumor vasculature, and antigens for accurate and efficient drug delivery to tumor cells. Furthermore, the development of multifunctional nanoparticle drug delivery systems, such as folate-modified chitosan micelles, enables passive tumor targeting through enhanced permeability and retention effects, leading to increased drug accumulation in tumor areas while minimizing toxic effects on normal tissues. Mimicking malaria-infected erythrocytes, nanoparticles functionalized with VAR2CSA specifically target oncofetal chondroitin sulfate expressed by many cancers, showcasing a promising strategy for tumor-specific drug delivery.
How combine Biomaterial-based DDS and cancer immunotherapy?5 answersBiomaterial-based drug delivery systems (DDS) can enhance cancer immunotherapy by improving drug concentration, localization, and controlled release. These systems offer advantages in delivering drugs, cancer antigens, and immune cells effectively. For instance, engineered biomaterials have been utilized to enhance dendritic cell (DC)-mediated immunotherapy by optimizing DC functions through targeting modifications, size, shape, and surface properties. In the context of cancer vaccines, in situ vaccines generated from tumors have shown promise in eliciting potent immune responses. Biomaterials-based in situ cancer vaccines have been successful in synergizing antigens and adjuvants, addressing delivery challenges, and manipulating immune cells effectively. Additionally, injectable biomaterial platforms like SynerGel have demonstrated improved intratumoral drug delivery for cancer immunotherapy, leading to enhanced treatment efficacy in preclinical models.
Is there any research articles using same biopolymer nanocarrier for different anticancer drugs?5 answersPolymeric nanocarriers have been extensively researched for the delivery of different anticancer drugs. Various types of nanocarriers, such as organic-inorganic-hybrid NPs and polymeric NPs (PNPs), have been developed for this purpose. Biopolymer nanovehicles have been explored for the oral delivery of natural anticancer agents (NAAs). Polymer colloids, including micelles, liposomes, emulsions, cationic carriers, and hydrogels, have also been used as drug delivery agents in cancer treatment. Additionally, polymeric nanoparticles, such as micelles, liposomes, dendrimers, polymersomes, hydrogels, and metal-organic frameworks, have been investigated for their potential in cancer drug delivery. Polymers derived from both natural and synthetic sources, such as poly(lactic-co-glycolic acid) (PLGA), polycaprolactone (PCL), and polyethylene glycol (PEG), have shown promise as carriers for different anticancer drugs. Therefore, there are several research articles that have utilized the same biopolymer nanocarrier for different anticancer drugs.
How can nano medicine benifite for cancer patients?5 answersNanomedicine offers several benefits for cancer patients. It allows for the development of innovative drug carriers that have greater efficacy and fewer side effects compared to conventional therapies. Nanoparticles can be engineered to target specific tumor sites, improving drug delivery and reducing toxicity. Nanoparticle-based platforms can modulate the biodistribution and accumulation of chemotherapeutic drugs, enhancing their efficacy while reducing their toxicity. Nanoparticles can also be used for cancer diagnosis, providing noninvasive and sensitive methods for early and precise detection. Additionally, the combination of nanoparticles with photosensitizers enables targeted photodynamic therapy, improving treatment outcomes with reduced side effects. Nanoparticles can also be used to deliver other cancer treatments, such as radiation therapy and gene therapy, directly to the tumor site, increasing treatment effectiveness while minimizing damage to healthy tissues. Overall, nanomedicine holds great promise for improving cancer diagnosis and treatment outcomes.
What are the most promising drug delivery approaches of nanomedicine in cancer research?5 answersNanomedicine offers promising drug delivery approaches in cancer research. Polymeric micelles, such as those made of stimuli-sensitive polymers, are being investigated as nanomedicines for targeted cancer treatment. Quantum dots (QDs) and nanoparticles (NPs) are emerging as bioimaging probes for cancer nanomedicine, enabling deep-tissue bimodal imaging and therapy. Metallic nanostructures and core-shell nanocomposites are being explored for combined photothermal-chemo or radio-chemo therapy. Nanoparticle drug formulations, particularly those utilizing hydrogel micropatterning, show promise in accurately assessing drug uptake and efficacy in different cancer cell fractions. Poly lactic-co-glycolic acid (PLGA) NPs, with their controlled and sustained drug release, are being used in passive and active drug delivery systems for efficient cancer therapy. Overall, nanomedicine approaches aim to achieve targeted drug delivery to the tumor site, improving therapeutic efficacy and reducing side effects.
How can nanoparticles be used to treat cancer?3 answersNanoparticles can be used to treat cancer through various mechanisms. Metallic nanoparticles such as palladium, gold, silver, and platinum have shown anticancer effects and can be functionalized with biomolecules to enhance their specificity and effectiveness against different types of cancer. Lanthanide-doped upconversion nanoparticles (UCNPs) can convert tissue-penetrating NIR light into visible and ultraviolet range, which can activate photosensitizers and regulate drug delivery processes for cancer therapy. Nanomedicine, including quantum dots, iron oxide NPs, PLGA NPs, dendrimer NPs, carbon nanotubes, liposomes, and gold NPs, has emerged as a promising approach to overcome therapeutic resistance, improve drug delivery, and target tumor cells in colorectal cancer. Nanoparticle-based delivery of anti-cancer drugs offers attractive features such as targeted and personalized treatment, prevention of tumor progression, and reduced side effects. Tumor cell membrane-coated nanoplatforms have been developed to evade immune system clearance and efficiently target tumor tissues for drug accumulation, providing a new strategy for precise and efficient chemotherapy of prostate cancer.

See what other people are reading

What are the Clinical and preclinical states of CAR T cell therapy in vivo for solid tumors cancer?
5 answers
Clinical and preclinical states of CAR T-cell therapy for solid tumors cancer are evolving. Challenges like antigen heterogeneity and immunosuppressive tumor microenvironment hinder CAR T-cell efficacy in solid tumors. Strategies to enhance CAR T-cell function include incorporating co-stimulatory domains, developing armored CAR-T cells, and exploring other immune cells like CAR-NK and CAR-M cells. Novel CAR T-cell products like YTB323 show enhanced in vivo expansion and antitumor activity in preclinical models, with promising safety and efficacy profiles in clinical trials for r/r DLBCL. To overcome limitations such as restricted trafficking, hypoxic TME, antigen escape, and exhaustion, researchers are advancing CAR designs and combining therapies to optimize clinical efficacy in solid tumors.
How does epigenetics affect cell Immunotherapy efficiency?
5 answers
Epigenetics plays a crucial role in influencing cell immunotherapy efficiency by modulating gene expression and chromatin architecture. Epigenetic alterations, such as DNA methylation and histone modifications, are common in cancer and can impact immune responses. Studies suggest that targeting epigenetic regulators can reverse immune escape mechanisms, potentially enhancing the effectiveness of immunotherapies. Furthermore, epigenetic therapies, when combined with immunotherapies, show promise in improving treatment outcomes for various cancers, including T-cell malignancies. Understanding the crosstalk between epigenetic modifications and immune responses is essential for identifying patients who will benefit from immunotherapy-based treatments and for developing novel therapeutic strategies.
What specific markers should be analyzed to assess immune cell activity in B2M knockout cultures?
5 answers
To assess immune cell activity in B2M knockout cultures, specific markers such as CD4, CD8, IL-2, TNF-alpha levels, and CD63 should be analyzed. CD4 and CD8 are crucial markers for T cell subsets. Additionally, IL-2 and TNF-alpha levels are indicative of immune response and rejection. CD63, a marker of exosomes, plays a role in mediating paracrine effects and preventing mitochondrial dysfunction and apoptosis in cardiomyocytes. These markers provide insights into the immune response, rejection, and functional outcomes in B2M knockout cultures, aiding in the evaluation of immune cell activity and the potential for immune rejection in these cultures.
What specific markers should be analyzed to assess immune cell activity in B2M knockout cultures in mice?
5 answers
To assess immune cell activity in B2M knockout cultures in mice, specific markers should be analyzed. These markers include MHC Class I molecules, immune subunits like LMP7, LMP2, and PA28, as well as regulators of proteasomes such as PA28 and PA700. Additionally, the expression of TDO2, a key enzyme in tryptophan metabolism, should be considered, especially in peritoneal macrophages where a reduced M1/M2 ratio was observed in TDO2 knockout mice. Furthermore, the deletion of β2-microglobulin (B2M) in human umbilical mesenchymal stem cells (UMSCs) can be assessed by examining the expression of HLA light chain β2-microglobulin (B2M) and its impact on immune rejection in mouse models. By analyzing these markers, researchers can gain insights into the immune response and activity in B2M knockout cultures in mice.
How does the expression of CD31/PECAM-1 and VE-Cadherin differ in B2M knockout mice compared to wild-type mice?
4 answers
In B2M knockout mice compared to wild-type mice, the expression of CD31/PECAM-1 and VE-Cadherin may exhibit significant differences. CD31/PECAM-1, an immunoglobulin superfamily member, plays a crucial role in lymphomas by facilitating survival and regulating extranodal spread of malignant cells. On the other hand, VE-Cadherin is essential for endothelial cell junction integrity and barrier function. The knockout of B2M could potentially impact the expression of these adhesion molecules, affecting vascular permeability and cellular responsiveness. Additionally, changes in gene expression variability in knockouts can influence phenotypic outcomes, potentially altering the expression levels of CD31/PECAM-1 and VE-Cadherin in B2M knockout mice. Further studies are needed to elucidate the specific alterations in CD31/PECAM-1 and VE-Cadherin expression in B2M knockout models.
Why are iPSCs not in human clinical trials?
5 answers
iPSCs are not currently in human clinical trials due to concerns regarding their immune response. Research has shown that iPSC-derived cells can be recognized and targeted by NK-cells, leading to cytotoxicity. This sensitivity to NK-cell activation, regardless of HLA-I status, highlights a potential risk in using iPSC-based therapies without considering the balance of ligands for NK-cell receptors. Additionally, the lack of inhibitory signals in iPSC-derivatives compared to parental cells may be due to their insufficient maturity, further emphasizing the need for caution in iPSC-based treatments. These immune responses and potential risks need to be thoroughly understood and addressed before iPSCs can advance to human clinical trials.
Does the hyperphosphorylation after infection within neurons differ between the sexes?
5 answers
Hyperphosphorylation after infection within neurons can differ based on the specific context of the infection. In prion-infected neurons, phosphorylation of protein kinase RNA-activated-like endoplasmic reticulum kinase (PERK) is enhanced, inducing endoplasmic reticulum stress but not fully activating the unfolded protein response. On the other hand, in the context of Alzheimer's disease, infections with Cdk5 inhibitory peptide selectively inhibit p25/Cdk5 activity, reducing aberrant tau phosphorylation in cortical neurons. Moreover, in the case of pneumonia-induced tau pathology, Ser-214 phosphorylation of tau is crucial for the cytotoxic properties of infection-induced oligomeric tau, impacting both lung and brain function. Therefore, the extent and consequences of hyperphosphorylation after infection within neurons can vary depending on the specific neurodegenerative disease context.
What are the potential side effects and limitations of current cancer inhibition techniques?
5 answers
Current cancer inhibition techniques, such as immunotherapy by checkpoint inhibition, have shown efficacy in various malignancies but come with limitations and potential side effects. Side effects of anti-cancer drugs include nephrogenic diabetes insipidus, thrombosis, protein-loss enteritis, heart failure, lactic acidosis, and severe diarrhea. Immunotherapy with immune checkpoint inhibitors can lead to toxicities and resistance, restricting effective responses in some patients. Additionally, there is a need for better biomarkers to identify patients who will benefit from checkpoint inhibition treatment. Combining locoregional therapies with immune checkpoint inhibitors is a promising approach to enhance therapeutic capabilities in oncology, but further research is required to optimize treatment strategies and minimize unnecessary side effects.
How does water kefir affect cancer cells by apoptosis?
5 answers
Water kefir, a probiotic beverage, exerts anticancer effects on cancer cells through apoptosis. Studies have shown that kefir induces apoptosis in cancer cells via a mitochondrial-dependent pathway, evidenced by upregulation of p53 expression, increased Bax/Bcl-2 ratio, decreased mitochondrial membrane potential (MMP) polarization, and caspase-3 activation. Additionally, kefir inhibits tumor proliferation by promoting cancer cell apoptosis, stimulating T helper cells and cytotoxic T cells, and displaying anti-inflammatory, antimetastatic, and antiangiogenic effects. The bioactive compounds in kefir, such as polysaccharides and peptides, have been identified as key players in inhibiting proliferation and inducing apoptosis in tumor cells. Furthermore, kefir has been found to induce cell cycle arrest and apoptosis in colorectal cancer cells, showcasing its potential as an anticancer agent.
What is the clinical outcome of CAR T cells therapy?
5 answers
CAR-T cell therapy has shown promising clinical outcomes in hematological malignancies. It targets antigens like CD19 and BCMA, inducing prolonged remissions with minimal long-term toxicities, potentially being curative for some patients. Notably, a single nucleotide polymorphism in the CD19 gene, encoding leucine or valine at position 174, influences treatment outcomes, with the minor allele L174 predicting a favorable response. CAR-T cells are custom-made for each patient, engineered to express chimeric proteins that recognize specific antigens on tumor cells, leading to robust anti-tumor responses. Despite heterogeneity in patient responses and safety concerns like cytokine release syndrome, ongoing research aims to identify factors affecting efficacy and improve long-term sustainability of this therapy. The success of CAR-T therapy in refractory B-cell malignancies has sparked hope for targeting other traditionally treatment-resistant cancers.
Papaya combined with probiotic have anticancer effect?
4 answers
Combining papaya with probiotics can indeed have anticancer effects. Papaya fermentation products have shown the ability to inhibit tumor growth, while probiotics have been documented to play a crucial role in preventing and treating various forms of cancer through microbiota and immunological modulation. Additionally, fermented papaya preparations have demonstrated chemotherapeutic properties by suppressing tumor growth and malignancy incidence. The synergistic effect of combining papaya and probiotics can potentially enhance their individual anticancer properties. This combination can offer advantages by targeting cancer cells directly, reducing the harmful effects of chemotherapy, and improving therapeutic impact. Further research is needed to explore the full potential of this combination in cancer treatment.