1,160 research outputs found

    Melanocortin 1 receptor targeted imaging of melanoma with gold nanocages and positron emission tomography

    Get PDF
    Purpose: Melanoma is a lethal skin cancer with unmet clinical needs for targeted imaging and therapy. Nanoscale materials conjugated with targeting components have shown great potential to improve tumor delivery efficiency while minimizing undesirable side effects in vivo. Herein, we proposed to develop targeted nanoparticles for melanoma theranostics. Method: In this work, gold nanocages (AuNCs) were conjugated with α-melanocyte-stimulating hormone (α-MSH) peptide and radiolabeled with 64Cu for melanocortin 1 receptor-(MC1R) targeted positron emission tomography (PET) in a mouse B16/F10 melanoma model. Results: Their controlled synthesis and surface chemistry enabled well-defined structure and radiolabeling efficiency. In vivo pharmacokinetic evaluation demonstrated comparable organ distribution between the targeted and nontargeted AuNCs. However, micro-PET/computed tomography (CT) imaging demonstrated specific and improved tumor accumulation via MC1R-mediated delivery. By increasing the coverage density of α-MSH peptide on AuNCs, the tumor delivery efficiency was improved. Conclusion: The controlled synthesis, sensitive PET imaging, and optimal tumor targeting suggested the potential of targeted AuNCs for melanoma theranostics. </jats:sec

    iRGD peptide conjugation potentiates intraperitoneal tumor delivery of paclitaxel with polymersomes

    Get PDF
    Polymersomes are versatile nanoscale vesicles that can be used for cytoplasmic delivery of payloads. Recently, we demonstrated that pH-sensitive polymersomes exhibit an intrinsic selectivity towards intraperitoneal tumor lesions. A tumor homing peptide, iRGD, harbors a cryptic C-end Rule (CendR) motif that is responsible for neuropilin-1 (NRP-1) binding and for triggering extravasation and tumor penetration of the peptide. iRGD functionalization increases tumor selectivity and therapeutic efficacy of systemic drug-loaded nanoparticles in many tumor models. Here we studied whether intraperitoneally administered paclitaxel-loaded iRGD-polymersomes show improved efficacy in the treatment of peritoneal carcinomatosis. First, we demonstrated that the pH-sensitive polymersomes functionalized with RPARPAR (a prototypic CendR peptide) or iRGD internalize in the cells that express NRP-1, and that internalized polymersomes release their cargo inside the cytosol. CendR-targeted polymersomes loaded with paclitaxel were more cytotoxic on NRP-1-positive cells than on NRP-1-negative cells. In mice bearing peritoneal tumors of gastric (MKN-45P) or colon (CT26) origin, intraperitoneally administered RPARPAR and iRGD-polymersomes showed higher tumor-selective accumulation and penetration than untargeted polymersomes. Finally, iRGD-polymersomes loaded with paclitaxel showed improved efficacy in peritoneal tumor growth inhibition and in suppression of local dissemination compared to the pristine paclitaxel-polymersomes or Abraxane. Our study demonstrates that iRGD-functionalization improves efficacy of paclitaxel-polymersomes for intraperitoneal treatment of peritoneal carcinomatosis

    Icp-Ms Analysis of Lanthanide-Doped Nanoparticles: A Quantitative and Multiplexing Approach to Investigate Biodistribution, Blood Clearance, and Targeting

    Get PDF
    The rapidly progressing field of nanotechnology promises to revolutionize healthcare in the 21st century, with applications in the prevention, diagnosis, and treatment of a wide range of diseases. However, before nanoparticulate agents can be brought into clinical use, they must first be developed, optimized, and evaluated in animal models. In the typical pre-clinical paradigm, almost all of the optimization is done at the in vitro level, with only a few select agents reaching the level of animal studies. Since only one experimental nanoparticle formulation can be investigated in a single animal, and in vivo experiments have relatively higher complexity, cost, and time requirements, it is not feasible to evaluate a very large number of agents at the in vivo stage. A major drawback of this approach, however, is that in vitro assays do not always accurately predict how a nanoparticle will perform in animal studies. Therefore, a method that allows many agents to be evaluated in a single animal subject would allow for much more efficient and predictive optimization of nanoparticles. We have found that by incorporating lanthanide tracer metals into nanoparticle formulations, we are successfully able to use inductively coupled plasma mass spectrometry (ICP-MS) to quantitatively determine a nanoparticle\u27s blood clearance kinetics, biodistribution, and tumor delivery. This approach was applied to evaluate both passive and active tumor targeting, as well as metabolically directed targeting of nanoparticles to low pH tumor microenvironments. Importantly, we found that these in vivo measurements could be made for many nanoparticle formulations simultaneously, in single animals, due to the high-order multiplexing capability of mass spectrometry. This approach allowed for efficient and reproducible comparison of performance between different nanoparticle formulations, by eliminating the effects of subject-to-subject variability. In the future, we envision that this higher-throughput evaluation of agents at the in vivo level, using ICP-MS multiplex analysis, will constitute a powerful tool to accelerate pre-clinical evaluation of nanoparticles in animal models

    Developing pharmacokinetic models for nano drug delivery systems

    Get PDF
    Trabalho Final de Mestrado Integrado, Ciências Farmacêuticas, 2021, Universidade de Lisboa, Faculdade de Farmácia.A área dos nanomedicamentos é interdisciplinar e complexa com fontes de literatura terciárias, sobre a forma de manuais, emergentes desde os 2010 e, ainda assim, os processos que sustentam a farmacocinética e a farmacodinâmica de nanomedicamentos ainda não estão totalmente caracterizados. O objetivo desta monografia é apresentar, para os indivíduos que podem ser relativamente novos na área de nanomedicamentos, as propriedades farmacocinéticas de nanopartículas, as abordagens na modelação farmacocinética, e demonstrar a aplicação destes princípios em exemplos tanto de investigação fundamental, quanto no desenvolvimento e otimização bio galénica de nanomedicamentos. Aqui são descritas as etapas farmacocinéticas de absorção, distribuição, metabolização e eliminação referentes a nanomedicamentos, com realce nos aspetos que distinguem estes processos daquilo que é observado quando se trata de medicamentos “convencionais”. É também fornecida uma discussão sobre conceitos essenciais necessários para discussão de modelação farmacocinética usados nas abordagens compartimentais, mecanísticas, e baseadas na fisiologia. Diversos assuntos tangentes como corrente interesse na área de oncologia, extrapolação interespécies em estudos pré-clínicos e aspetos regulamentares associados são também brevemente abordados. Esta monografia foi realizada com base nas publicações disponíveis nas bases de dados de PubMed e Science Direct até ao mês de setembro do ano 2021. Este trabalho não é único e assemelha-se as revisões de Moss D. M. e Siccardi M., de Glassman P. M. e Muzakantov V. R., ou de Yuan D. et al quanto a organização bem como aos conteúdos.(1–3) A farmacocinética que descreve os medicamentos “convencionais” baseados na distribuição de substâncias ativas começa apenas quando as etapas finais de libertação e degradação das nanopartículas já começam a ocorrer. A existência simultânea de entidades particuladas e moleculares complica a descrição, otimização, desenvolvimento e avaliação regulamentar de novas formulações de nanomedicamentos. Isto, juntamente com a falta de técnicas analíticas adequadas para a quantificação de nanopartículas em meios biológicos, torna os estudos de modelação farmacocinética de nanomedicamentos um desafio.Nanomedicines are a complex and highly interdisciplinary field with recently emerging Textbooks as tertiary literature sources since 2010s, and yet the processes that underpin the pharmacokinetics and pharmacodynamics of nano drug delivery systems are not fully characterized. The aim of this monograph is to introduce the pharmacokinetic dispositions, pharmacokinetic modelling approaches, and to demonstrate application of these principles in examples of both basic research and NDDS development to individuals who may be relatively new to the field of nanomedicine. In this monograph are described the pharmacokinetic steps of absorption, distribution, metabolization and elimination particular to nano drug delivery systems, primarily focusing aspects that distinguish NDDS from “conventional” drugs. A description of essential concepts necessary for discussions of PK modelling in compartmental, mechanistic, and physiology-based approaches are also provided. Various related topics including growing interest in cancer therapy, interspecies extrapolation in pre-clinical study settings, and reglementary affairs related to NDDSs are also briefly addressed. Writing of this monograph was conducted after browsing information available in the PubMed and Science Direct databases up to September 2021. This work is not unique and resembles the reviews by Moss D. M. and Siccardi M., Glassman P. M. and Muzakantov V. R., and Yuan D. et al, in their structure, subject and contents.(1–3) Pharmacokinetics that describes small molecule active substances, begin only when the final steps of nanoparticles fate of release and degradation had begun. Simultaneous existence of both particulate and molecular entities complicates the description, optimization, development, and regulatory assessment of new nano formulations. This together with the lack of appropriate analytical techniques for nanoparticle quantification in biologic media makes pharmacokinetic modelling studies of NDDSs challenging

    64Cu-MM-302 Positron Emission Tomography Quantifies Variability of Enhanced Permeability and Retention of Nanoparticles in Relation to Treatment Response in Patients with Metastatic Breast Cancer

    Get PDF
    Purpose: Therapeutic nanoparticles are designed to deliver their drug payloads through enhanced permeability and retention (EPR) in solid tumors. The extent of EPR and its variability in human tumors is highly debated and has been proposed as an explanation for variable responses to therapeutic nanoparticles in clinical studies. Experimental Design: We assessed the EPR effect in patients using a 64Cu-labeled nanoparticle, 64Cu-MM-302 (64Cu-labeled HER2-targeted PEGylated liposomal doxorubicin), and imaging by PET/CT. Nineteen patients with HER2-positive metastatic breast cancer underwent 2 to 3 PET/CT scans postadministration of 64Cu-MM-302 as part of a clinical trial of MM-302 plus trastuzumab with and without cyclophosphamide (NCT01304797). Results: Significant background uptake of 64Cu-MM-302 was observed in liver and spleen. Tumor accumulation of 64Cu-MM-302 at 24 to 48 hours varied 35-fold (0.52–18.5 %ID/kg), including deposition in bone and brain lesions, and was independent of systemic plasma exposure. Computational analysis quantified rates of deposition and washout, indicating peak liposome deposition at 24 to 48 hours. Patients were classified on the basis of 64Cu-MM-302 lesion deposition using a cut-off point that is comparable with a response threshold in preclinical studies. In a retrospective exploratory analysis of patient outcomes relating to drug levels in tumor lesions, high 64Cu-MM-302 deposition was associated with more favorable treatment outcomes (HR = 0.42). Conclusions: These findings provide important evidence and quantification of the EPR effect in human metastatic tumors and support imaging nanoparticle deposition in tumors as a potential means to identify patients well suited for treatment with therapeutic nanoparticles

    Minibeam radiation therapy enhanced tumor delivery of PEGylated liposomal doxorubicin in a triple-negative breast cancer mouse model

    Get PDF
    Background: Minibeam radiation therapy is an experimental radiation therapy utilizing an array of parallel submillimeter planar X-ray beams. In preclinical studies, minibeam radiation therapy has been shown to eradicate tumors and cause significantly less damage to normal tissue compared to equivalent radiation doses delivered by conventional broadbeam radiation therapy, where radiation dose is uniformly distributed. Methods: Expanding on prior studies that suggested minibeam radiation therapy increased perfusion in tumors, we compared a single fraction of minibeam radiation therapy (peak dose:valley dose of 28 Gy:2.1 Gy and 100 Gy:7.5 Gy) and broadbeam radiation therapy (7 Gy) in their ability to enhance tumor delivery of PEGylated liposomal doxorubicin and alter the tumor microenvironment in a murine tumor model. Plasma and tumor pharmacokinetic studies of PEGylated liposomal doxorubicin and tumor microenvironment profiling were performed in a genetically engineered mouse model of claudin-low triple-negative breast cancer (T11). Results: Minibeam radiation therapy (28 Gy) and broadbeam radiation therapy (7 Gy) increased PEGylated liposomal doxorubicin tumor delivery by 7.1-fold and 2.7-fold, respectively, compared to PEGylated liposomal doxorubicin alone, without altering the plasma disposition. The enhanced tumor delivery of PEGylated liposomal doxorubicin by minibeam radiation therapy is consistent after repeated dosing, is associated with changes in tumor macrophages but not collagen or angiogenesis, and nontoxic to local tissues. Our study indicated that the minibeam radiation therapy’s ability to enhance the drug delivery decreases from 28 to 100 Gy peak dose. Discussion: Our studies suggest that low-dose minibeam radiation therapy is a safe and effective method to significantly enhance the tumor delivery of nanoparticle agents

    Enhanced tumor delivery and antitumor response of doxorubicin loaded albumin nanoparticles formulated based on a Schiff base

    Get PDF
    A novel method was developed here to prepare albumin-based nanoparticles (NPs) for improving the therapeutic and safety profiles of chemotherapeutic agents. This approach involved crosslinking bovine serum albumin (BSA) using a Schiff base-containing vanillin, into NPs and loading doxorubicin (DOX) into the NPs by incubation. The resultant NPs (DOX-BSA-V-NPs) displayed a particle size of 100.5±1.3 nm with a zeta potential of −23.05±1.45 mV and also showed high drug-loading efficiency and excellent stability with respect to storage and temperature. The encapsulation of DOX into the BSA-V-NPs was confirmed by dynamic scanning calorimetry and Raman spectroscopy. DOX-BSA-V-NPs exhibited a significantly faster DOX release at pH 6.5 than pH 7.4, as well as in a solution with a higher glutathione concentration. In vitro studies showed that the cellular uptake of DOX-BSA-V-NPs was time-dependent, concentration-dependent, and faster than free DOX, while the cytotoxicity of DOX-BSA-V-NPs (IC50 value of 3.693 μg/mL) was superior to free DOX (IC50 value of 4.007 μg/mL). More importantly, DOX-BSA-V-NPs showed a longer mean survival time of 24.83 days, a higher tumor inhibition rate of 56.66%, and a decreased distribution in the heart than other DOX formulations in animal studies using a tumor xenograft model. Thus, the vanillin-based albumin NPs were shown here to be a promising carrier for tumor-targeted delivery of chemotherapeutic agents and, thus, should be further studied
    corecore