Pub Date : 2023-01-01DOI: 10.1016/j.nano.2022.102613
C.G. Anjali Das M.Sc, V. Ganesh Kumar Ph.D, T. Stalin Dhas Ph.D, V. Karthick Ph.D, C.M. Vineeth Kumar M.Sc
The current challenges in cancer treatment using conventional therapies have made the emergence of nanotechnology with more advancements. The exponential growth of nanoscience has drawn to develop nanomaterials (NMs) with therapeutic activities. NMs have enormous potential in cancer treatment by altering the drug toxicity profile. Nanoparticles (NPs) with enhanced surface characteristics can diffuse more easily inside tumor cells, thus delivering an optimal concentration of drugs at tumor site while reducing the toxicity. Cancer cells can be targeted with greater affinity by utilizing NMs with tumor specific constituents. Furthermore, it bypasses the bottlenecks of indiscriminate biodistribution of the antitumor agent and high administration dosage. Here, we focus on the recent advances on the use of various nanomaterials for cancer treatment, including targeting cancer cell surfaces, tumor microenvironment (TME), organelles, and their mechanism of action. The paradigm shift in cancer management is achieved through the implementation of anticancer drug delivery using nano routes.
{"title":"Nanomaterials in anticancer applications and their mechanism of action - A review","authors":"C.G. Anjali Das M.Sc, V. Ganesh Kumar Ph.D, T. Stalin Dhas Ph.D, V. Karthick Ph.D, C.M. Vineeth Kumar M.Sc","doi":"10.1016/j.nano.2022.102613","DOIUrl":"https://doi.org/10.1016/j.nano.2022.102613","url":null,"abstract":"<div><p><span>The current challenges in cancer treatment using conventional therapies<span><span> have made the emergence of nanotechnology with more advancements. The exponential growth of nanoscience has drawn to develop </span>nanomaterials (NMs) with therapeutic activities. NMs have enormous potential in cancer treatment by altering the </span></span>drug toxicity<span> profile. Nanoparticles<span><span> (NPs) with enhanced surface characteristics can diffuse more easily inside tumor cells, thus delivering an optimal concentration of drugs<span> at tumor site while reducing the toxicity. Cancer cells can be targeted with greater affinity by utilizing NMs with </span></span>tumor specific<span><span> constituents. Furthermore, it bypasses the bottlenecks of indiscriminate biodistribution<span> of the antitumor agent and high administration dosage. Here, we focus on the recent advances on the use of various nanomaterials for cancer treatment, including targeting cancer cell surfaces, tumor </span></span>microenvironment (TME), organelles, and their mechanism of action. The paradigm shift in cancer management is achieved through the implementation of anticancer drug delivery using nano routes.</span></span></span></p></div>","PeriodicalId":396,"journal":{"name":"Nanomedicine: Nanotechnology, Biology and Medicine","volume":"47 ","pages":"Article 102613"},"PeriodicalIF":5.5,"publicationDate":"2023-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"1567242","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2023-01-01DOI: 10.1016/j.nano.2022.102617
Jiahui Wang MS , Liang Li PhD , Yanbo Li MD , Liangsheng Liu MD , Junnan Li MS , Xiaokang Li MS , Ying Zhu MS , Xuening Zhang PhD , Hong Lu PhD
Prostate-specific membrane antigen (PSMA) is a prominent biomarker for prostate cancer (PCa) diagnosis. Safe contrast agents able to render the expression and distribution of PSMA would facilitate early accurate screening and prognostic prediction of PCa. However, current Gd-containing nanoparticles are often limited by nonspecific redistribution in mononuclear phagocyte system (MPS) and inadequate perfusion to target sites. Besides, intrinsic defects of magnetic resonance (MR) equipment also hamper their use for precisely depicting PSMA details. Herein, we devised a novel noninvasive MR/CT/NIRF multimodal contrast agent (AGGP) coordinated to a high-affinity PSMA ligand (PSMA1) to specifically detect and quantify PSMA expression in PCa lesions, which exhibited formidable tripe-modal signal augments, preferential PSMA targeting, effective MPS escaping and profitable renal-clearable behavior in living mice. Biocompatibility and histopathological studies substantiated high security of AGGP in vivo, opening the door to future opportunities for improving early-stage PCa detection and clinical implementation of more effective multifunctional nanotherapeutics.
{"title":"PSMA1-mediated ultrasmall gold nanoparticles facilitate tumor targeting and MR/CT/NIRF multimodal detection of early-stage prostate cancer","authors":"Jiahui Wang MS , Liang Li PhD , Yanbo Li MD , Liangsheng Liu MD , Junnan Li MS , Xiaokang Li MS , Ying Zhu MS , Xuening Zhang PhD , Hong Lu PhD","doi":"10.1016/j.nano.2022.102617","DOIUrl":"https://doi.org/10.1016/j.nano.2022.102617","url":null,"abstract":"<div><p>Prostate-specific membrane antigen (PSMA) is a prominent biomarker for prostate cancer (PCa) diagnosis. Safe contrast agents able to render the expression and distribution of PSMA would facilitate early accurate screening and prognostic prediction of PCa. However, current Gd-containing nanoparticles are often limited by nonspecific redistribution in mononuclear phagocyte system (MPS) and inadequate perfusion to target sites. Besides, intrinsic defects of magnetic resonance (MR) equipment also hamper their use for precisely depicting PSMA details. Herein, we devised a novel noninvasive MR/CT/NIRF multimodal contrast agent (AGGP) coordinated to a high-affinity PSMA ligand (PSMA1) to specifically detect and quantify PSMA expression in PCa lesions, which exhibited formidable tripe-modal signal augments, preferential PSMA targeting, effective MPS escaping and profitable renal-clearable behavior in living mice. Biocompatibility and histopathological studies substantiated high security of AGGP <em>in vivo</em>, opening the door to future opportunities for improving early-stage PCa detection and clinical implementation of more effective multifunctional nanotherapeutics.</p></div>","PeriodicalId":396,"journal":{"name":"Nanomedicine: Nanotechnology, Biology and Medicine","volume":"47 ","pages":"Article 102617"},"PeriodicalIF":5.5,"publicationDate":"2023-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"3209045","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2023-01-01DOI: 10.1016/j.nano.2022.102618
Jing-jing Ji PhD , Shang-yu Chen PhD , Zi-wei Yang MSc , Rui Zhang PhD , Ling-lin Qian PhD , Yu Jiang PhD , Jia-qi Guo PhD , Ya Wu PhD , Qu-li Fan PhD , Yu-yu Yao PhD , Peng-fei Sun PhD
Ferroptosis plays an important role in ischemia-reperfusion (I/R)-induced cardiac injury and there are many defects in current targeted delivery of miRNAs for the treatment of ferroptosis. We herein report a unique hydrogel (Gel) that can be triggered by a near-infrared-II (NIR-II) light with deep tissue penetration and biocompatible maximum permissible exposure (MPE) value for in situ treatment after I/R. The mir-196c-3p mimic (mimics) and photothermal nanoparticles (BTN) were co-encapsulated in an injectable Gel (mimics + Gel/BTN) with NIR-II light-triggered release. Using 1064 nm light irradiation, local microenvironment photothermal-triggered on-demand noninvasive controllable delivery of miRNA was achieved, aiming to inhibit I/R-induced ferroptosis. Consequently, declined ferroptosis in cardiomyocytes and improved cardiac function, survival rate in rats was achieved through the controlled release of Gel/BTN mimics in I/R model to simultaneously inhibit ferroptosis hub genes NOX4, P53, and LOX expression.
{"title":"Delivery of Mir-196c-3p with NIR-II light-triggered gel attenuates cardiomyocyte ferroptosis in cardiac ischemia-reperfusion injury","authors":"Jing-jing Ji PhD , Shang-yu Chen PhD , Zi-wei Yang MSc , Rui Zhang PhD , Ling-lin Qian PhD , Yu Jiang PhD , Jia-qi Guo PhD , Ya Wu PhD , Qu-li Fan PhD , Yu-yu Yao PhD , Peng-fei Sun PhD","doi":"10.1016/j.nano.2022.102618","DOIUrl":"https://doi.org/10.1016/j.nano.2022.102618","url":null,"abstract":"<div><p>Ferroptosis plays an important role in ischemia-reperfusion (I/R)-induced cardiac injury and there are many defects in current targeted delivery of miRNAs for the treatment of ferroptosis. We herein report a unique hydrogel (Gel) that can be triggered by a near-infrared-II (NIR-II) light with deep tissue penetration and biocompatible maximum permissible exposure (MPE) value for <em>in situ</em> treatment after I/R. The mir-196c-3p mimic (mimics) and photothermal nanoparticles (BTN) were co-encapsulated in an injectable Gel (mimics + Gel/BTN) with NIR-II light-triggered release. Using 1064 nm light irradiation, local microenvironment photothermal-triggered on-demand noninvasive controllable delivery of miRNA was achieved, aiming to inhibit I/R-induced ferroptosis. Consequently, declined ferroptosis in cardiomyocytes and improved cardiac function, survival rate in rats was achieved through the controlled release of Gel/BTN mimics in I/R model to simultaneously inhibit ferroptosis hub genes NOX4, P53, and LOX expression.</p></div>","PeriodicalId":396,"journal":{"name":"Nanomedicine: Nanotechnology, Biology and Medicine","volume":"47 ","pages":"Article 102618"},"PeriodicalIF":5.5,"publicationDate":"2023-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"1567241","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Emerging clinically required α-synuclein (α-syn) inhibitor which acts as a neuroprotective nanocomposite drug is in increased demand as a patient-safe central nervous system therapeutic. This inhibitor is intended to chemically engineer graphene quantum dot (GQD) with blue luminescence, and stands to be a potential cure for Parkinson's disease. It has been theorized that α-syn aggregation is a critical step in the development of Parkinson's. Hence narrow the target by α-syn inhibition, through chemically synthesize methyl N-allyl N-benzoylmethioninate (MABM) and functionally engineer the surface of GQD to target the brain delivery on C57BL/6 mice. Spectroscopic and simulation studies confirm defibrillation through the interaction between N-terminal amino acids and MABM-GQD nanoparticles, which makes nontoxic α-syn. Therefore, this drug's ability to cross the blood-brain barrier in vitro functionally prevents neuronal loss in neuroblastoma cells. Thus, in vivo cerebral blood flow analysis using magnetic resonance imaging illustrates, how this nanocomposite can possibly treat Parkinson's.
{"title":"Engineered graphene quantum dot nanocomposite triggers α-synuclein defibrillation: Therapeutics against Parkinson's disease","authors":"Poonkuzhali Kaliyaperumal , Seenivasagan Renganathan , Karthika Arumugam , Bukola Rhoda Aremu","doi":"10.1016/j.nano.2022.102608","DOIUrl":"https://doi.org/10.1016/j.nano.2022.102608","url":null,"abstract":"<div><p><span><span><span>Emerging clinically required α-synuclein (α-syn) inhibitor which acts as a neuroprotective<span><span> nanocomposite </span>drug is in increased demand as a patient-safe </span></span>central nervous system<span> therapeutic. This inhibitor is intended to chemically engineer graphene quantum dot (GQD) with blue luminescence, and stands to be a potential cure for </span></span>Parkinson's disease. It has been theorized that α-syn aggregation is a critical step in the development of Parkinson's. Hence narrow the target by α-syn inhibition, through chemically synthesize methyl </span><em>N</em>-allyl <em>N</em><span>-benzoylmethioninate (MABM) and functionally engineer the surface of GQD to target the brain delivery on C57BL/6 mice. Spectroscopic and simulation studies confirm defibrillation<span> through the interaction between N-terminal amino acids<span> and MABM-GQD nanoparticles<span>, which makes nontoxic α-syn. Therefore, this drug's ability to cross the blood-brain barrier in vitro functionally prevents neuronal loss in neuroblastoma cells. Thus, in vivo cerebral blood flow analysis using magnetic resonance imaging illustrates, how this nanocomposite can possibly treat Parkinson's.</span></span></span></span></p></div>","PeriodicalId":396,"journal":{"name":"Nanomedicine: Nanotechnology, Biology and Medicine","volume":"47 ","pages":"Article 102608"},"PeriodicalIF":5.5,"publicationDate":"2023-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"3342726","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Combination chemotherapy with systemic administration of drugs in their free form can be challenging due to non-synchronized pharmacokinetics and sub-optimal tumor accumulation. The present study investigates a PLA-based block copolymeric nanocarrier for the co-delivery of navitoclax and decitabine (NAV/DCB NPs) for combination cancer therapy. NAV/DCB NPs exhibited potent in vitro synergistic cytotoxicity in both acute myeloid leukemia and breast cancer cell lines. Biodistribution studies of NAV/DCB NPs in tumor bearing mice, showed significant drug accumulation in tumor tissue and detectable quantities in plasma even after 48 h. Good hemocompatibility with reduced in vivo platelet toxicity indicated that encapsulation in PLA-based nanocarrier helped ameliorate navitoclax associated thrombocytopenia. In vivo biological activity of NAV/DCB NPs evaluated in xenograft AML and syngeneic breast cancer model, demonstrated potent tumor growth inhibition efficacy. PLA-based NAV/DCB dual NPs present a novel, safe and effective nanoformulation for combination cancer therapy in both solid tumors and hematologic malignancies.
{"title":"Polylactic acid based polymeric nanoparticle mediated co-delivery of navitoclax and decitabine for cancer therapy","authors":"Neha Mehrotra PhD , Mohd Anees M.Sc , Sachchidanand Tiwari M.Tech , Surender Kharbanda PhD , Harpal Singh PhD","doi":"10.1016/j.nano.2022.102627","DOIUrl":"https://doi.org/10.1016/j.nano.2022.102627","url":null,"abstract":"<div><p><span><span><span><span>Combination chemotherapy<span> with systemic administration of </span></span>drugs in their free form can be challenging due to non-synchronized </span>pharmacokinetics and sub-optimal tumor accumulation. The present study investigates a PLA-based block copolymeric </span>nanocarrier<span> for the co-delivery of navitoclax and </span></span>decitabine<span><span><span> (NAV/DCB NPs) for combination cancer therapy. NAV/DCB NPs exhibited potent in vitro synergistic cytotoxicity in both acute myeloid leukemia<span> and breast cancer cell lines<span>. Biodistribution studies of NAV/DCB NPs in tumor bearing mice, showed significant drug accumulation in tumor tissue and detectable quantities in plasma even after 48 h. Good </span></span></span>hemocompatibility<span><span> with reduced in vivo platelet toxicity indicated that encapsulation in PLA-based nanocarrier helped ameliorate navitoclax associated thrombocytopenia<span>. In vivo biological activity of NAV/DCB NPs evaluated in </span></span>xenograft<span> AML and </span></span></span>syngeneic<span> breast cancer model, demonstrated potent tumor growth inhibition efficacy. PLA-based NAV/DCB dual NPs present a novel, safe and effective nanoformulation for combination cancer therapy in both solid tumors<span> and hematologic malignancies.</span></span></span></p></div>","PeriodicalId":396,"journal":{"name":"Nanomedicine: Nanotechnology, Biology and Medicine","volume":"47 ","pages":"Article 102627"},"PeriodicalIF":5.5,"publicationDate":"2023-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"3457446","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2023-01-01DOI: 10.1016/j.nano.2022.102615
Rong Ma PhD , Xiaohui Tang MS , Mei Wang PhD , Zhong Du MS , Shuang Chen MS , Youqiang Heng MS , Lijun Zhu MS , Nuernisha Alifu PhD , Xueliang Zhang PhD , Cailing Ma PhD
Cervical diseases such as lymph node disease and tubal obstruction have threatened women's health. However, the traditional diagnostic methods still have shortcomings. NIR-II fluorescence imaging with advantages of low scattering, negligible autofluorescence, and high spatial resolution could be an ideal option. To obtain high quality NIR-II fluorescence imaging, selecting appropriate nanoprobes becomes the important issue. As a small molecular photothermal agent, extensive applications of ICG are rather limited because of its drawbacks. Herein, natural silk fibroin (SF) was synthesized and encapsulated ICG molecules to form SF@ICG nanoparticles (NPs). After detailed analysis, SF@ICG NPs showed excellent stability and long circulation time, as well as strong NIR-II fluorescence emission, well photo-stability, biocompatibility and well photothermal property under 808 nm laser irradiation. Furthermore, SF@ICG NPs were utilized for NIR-II fluorescence imaging of lymph node/lymphangiography and angiography of fallopian tubes. The process of fallopian tubes could be detected with high resolution and high sensitivity.
{"title":"Clinical indocyanine green-based silk fibroin theranostic nanoprobes for in vivo NIR-I/II fluorescence imaging of cervical diseases","authors":"Rong Ma PhD , Xiaohui Tang MS , Mei Wang PhD , Zhong Du MS , Shuang Chen MS , Youqiang Heng MS , Lijun Zhu MS , Nuernisha Alifu PhD , Xueliang Zhang PhD , Cailing Ma PhD","doi":"10.1016/j.nano.2022.102615","DOIUrl":"https://doi.org/10.1016/j.nano.2022.102615","url":null,"abstract":"<div><p>Cervical diseases such as lymph node disease and tubal obstruction have threatened women's health. However, the traditional diagnostic methods still have shortcomings. NIR-II fluorescence imaging with advantages of low scattering, negligible autofluorescence, and high spatial resolution could be an ideal option. To obtain high quality NIR-II fluorescence imaging, selecting appropriate nanoprobes becomes the important issue. As a small molecular photothermal agent, extensive applications of ICG are rather limited because of its drawbacks. Herein, natural silk fibroin (SF) was synthesized and encapsulated ICG molecules to form SF@ICG nanoparticles (NPs). After detailed analysis, SF@ICG NPs showed excellent stability and long circulation time, as well as strong NIR-II fluorescence emission, well photo-stability, biocompatibility and well photothermal property under 808 nm laser irradiation. Furthermore, SF@ICG NPs were utilized for NIR-II fluorescence imaging of lymph node/lymphangiography and angiography of fallopian tubes. The process of fallopian tubes could be detected with high resolution and high sensitivity.</p></div>","PeriodicalId":396,"journal":{"name":"Nanomedicine: Nanotechnology, Biology and Medicine","volume":"47 ","pages":"Article 102615"},"PeriodicalIF":5.5,"publicationDate":"2023-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"2891290","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
In a context of drug repurposing, pentamidine (PTM), an FDA-approved antiparasitic drug, has been proposed to reverse the splicing defects associated in myotonic dystrophy type 1 (DM1). However, clinical use of PTM is hinder by substantial toxicity, leading to find alternative delivery strategies. In this work we proposed hyaluronic acid-based nanoparticles as a novel encapsulation strategy to efficiently deliver PTM to skeletal muscles cells. In vitro studies on C2C12 myoblasts and myotubes showed an efficient nanoparticles' internalization with minimal toxicity. More interestingly, our findings evidenced for the first time the endosomal escape of hyaluronic acid-based nanocarriers. Ex vivo studies showed an efficient nanoparticles' internalization within skeletal muscle fibers. Finally, the therapeutic efficacy of PTM-loaded nanosystems to reduce the number of nuclear foci has been demonstrated in a novel DM1 in vitro model. So far, current data demonstrated the potency of hyaluronic acid-based nanosystems as efficient nanocarrier for delivering PTM into skeletal muscle and mitigate DM1 pathology.
{"title":"Repurposing pentamidine using hyaluronic acid-based nanocarriers for skeletal muscle treatment in myotonic dystrophy","authors":"Mathieu Repellin PhD , Flavia Carton PhD , Federico Boschi PhD , Mirco Galiè PhD , Massimiliano Perduca PhD , Laura Calderan PhD , Arnaud Jacquier PhD , Julien Carras MSc , Laurent Schaeffer PhD , Stéphanie Briançon PhD , Giovanna Lollo PhD , Manuela Malatesta MSc","doi":"10.1016/j.nano.2022.102623","DOIUrl":"https://doi.org/10.1016/j.nano.2022.102623","url":null,"abstract":"<div><p><span><span><span>In a context of drug repurposing, </span>pentamidine<span> (PTM), an FDA-approved antiparasitic drug, has been proposed to reverse the </span></span>splicing defects<span> associated in myotonic dystrophy type 1<span><span> (DM1). However, clinical use of PTM is hinder by substantial toxicity, leading to find alternative delivery strategies. In this work we proposed hyaluronic acid-based nanoparticles as a novel encapsulation strategy to efficiently deliver PTM to </span>skeletal muscles cells. </span></span></span><em>In vitro</em><span> studies on C2C12<span> myoblasts<span><span><span> and myotubes showed an efficient nanoparticles' </span>internalization with minimal toxicity. More interestingly, our findings evidenced for the first time the endosomal escape of hyaluronic acid-based </span>nanocarriers. </span></span></span><span><em>Ex vivo</em></span><span> studies showed an efficient nanoparticles' internalization within skeletal muscle<span> fibers. Finally, the therapeutic efficacy of PTM-loaded nanosystems to reduce the number of nuclear foci has been demonstrated in a novel DM1 </span></span><em>in vitro</em> model. So far, current data demonstrated the potency of hyaluronic acid-based nanosystems as efficient nanocarrier for delivering PTM into skeletal muscle and mitigate DM1 pathology.</p></div>","PeriodicalId":396,"journal":{"name":"Nanomedicine: Nanotechnology, Biology and Medicine","volume":"47 ","pages":"Article 102623"},"PeriodicalIF":5.5,"publicationDate":"2023-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"1567239","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2023-01-01DOI: 10.1016/j.nano.2022.102626
R. Rotem PhD , J.A. Bertolini PhD , L. Salvioni PhD , L. Barbieri MSc , M.A. Rizzuto PhD , V. Tinelli MSc , A. Gori PhD , S. Adams PhD , M. Colombo PhD , D. Prosperi PhD
The delivery of therapeutics across the cell membrane and into the cytoplasm is a major challenge that limits the development of new therapies. This challenge is compounded by the lack of a general assay for cytosolic delivery. Here we develop this assay based on the pro-fluorophore CrAsH-EDT2, and provide cytosolic penetration results for a variety of drug delivery agents (polyethyleneimine, poly-arginine, Ferritin, poly [maleic anhydride-alt-isobutene] grafted with dodecylamine, and cationic liposomes) into HeLa and T98G cells. Our results show that this method can be widely applicable to different cells and drug delivery agents, and yield statistically robust results. We later use this method to optimize and improve a model drug delivery agent's (Ferritin) cytosolic penetration.
{"title":"Direct quantification of cytosolic delivery of drug nanocarriers using FlAsH-EDT2","authors":"R. Rotem PhD , J.A. Bertolini PhD , L. Salvioni PhD , L. Barbieri MSc , M.A. Rizzuto PhD , V. Tinelli MSc , A. Gori PhD , S. Adams PhD , M. Colombo PhD , D. Prosperi PhD","doi":"10.1016/j.nano.2022.102626","DOIUrl":"https://doi.org/10.1016/j.nano.2022.102626","url":null,"abstract":"<div><p><span><span><span>The delivery of therapeutics across the cell membrane and into the cytoplasm is a major challenge that limits the development of new </span>therapies. This challenge is compounded by the lack of a general assay for cytosolic delivery. Here we develop this assay based on the pro-fluorophore CrAsH-EDT2, and provide cytosolic penetration results for a variety of drug delivery agents (polyethyleneimine, poly-arginine, </span>Ferritin, poly [maleic anhydride-</span><em>alt</em><span>-isobutene] grafted with dodecylamine, and cationic liposomes) into HeLa and T98G cells. Our results show that this method can be widely applicable to different cells and drug delivery agents, and yield statistically robust results. We later use this method to optimize and improve a model drug delivery agent's (Ferritin) cytosolic penetration.</span></p></div>","PeriodicalId":396,"journal":{"name":"Nanomedicine: Nanotechnology, Biology and Medicine","volume":"47 ","pages":"Article 102626"},"PeriodicalIF":5.5,"publicationDate":"2023-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"1567240","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2023-01-01DOI: 10.1016/j.nano.2022.102622
Qianyu Zhang PhD , Sai Li BS , Wen Wu PhD , Xuefeng Xia PhD , Jinqiang Zhang PhD
PASylation, which was recently reported as the conjugation of pharmacologically active compounds with polypeptide sequences mainly made of proline, alanine and serine, has been proposed as an alternative to PEGylation. In this study, we designed PAS-modified liposomes (PASylated liposomes) and studied the effect of the incorporation of PAS-lipid on the stability and pharmacokinetic properties of liposomes, and compared them both in vitro and in vivo to PEGylated liposomes. Results showed that PASylated liposomes modified with single-chained PAS-lipid C16-(PA3)7 (SC-PAS-Lip) showed comparable storage and serum stability to PEGylated liposomes (PEG-Lip), and a significantly decreased macrophage uptake compared with unmodified liposomes. SC-PAS-Lip displayed long circulating pharmacokinetic profile which was not impacted by the repeated administration of liposomes, and they were less likely to induce the production of anti-PEG IgM compared with PEGylated liposomes, presenting PASylation as an alternative liposome modification strategy to PEGylation.
{"title":"PASylation improves pharmacokinetic of liposomes and attenuates anti-PEG IgM production: An alternative to PEGylation","authors":"Qianyu Zhang PhD , Sai Li BS , Wen Wu PhD , Xuefeng Xia PhD , Jinqiang Zhang PhD","doi":"10.1016/j.nano.2022.102622","DOIUrl":"https://doi.org/10.1016/j.nano.2022.102622","url":null,"abstract":"<div><p>PASylation, which was recently reported as the conjugation of pharmacologically active compounds with polypeptide sequences mainly made of proline, alanine and serine, has been proposed as an alternative to PEGylation. In this study, we designed PAS-modified liposomes (PASylated liposomes) and studied the effect of the incorporation of PAS-lipid on the stability and pharmacokinetic properties of liposomes, and compared them both <em>in vitro</em> and <em>in vivo</em> to PEGylated liposomes. Results showed that PASylated liposomes modified with single-chained PAS-lipid C<sub>16</sub>-(PA<sub>3</sub>)<sub>7</sub> (SC-PAS-Lip) showed comparable storage and serum stability to PEGylated liposomes (PEG-Lip), and a significantly decreased macrophage uptake compared with unmodified liposomes. SC-PAS-Lip displayed long circulating pharmacokinetic profile which was not impacted by the repeated administration of liposomes, and they were less likely to induce the production of anti-PEG IgM compared with PEGylated liposomes, presenting PASylation as an alternative liposome modification strategy to PEGylation.</p></div>","PeriodicalId":396,"journal":{"name":"Nanomedicine: Nanotechnology, Biology and Medicine","volume":"47 ","pages":"Article 102622"},"PeriodicalIF":5.5,"publicationDate":"2023-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"1567238","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2023-01-01DOI: 10.1016/j.nano.2022.102609
Sahar Farhangi MSc , Elham Karimi MSc , Khosro Khajeh PhD , Saman Hosseinkhani PhD , Mohammad Javan PhD
Drug development for multiple sclerosis (MS) clinical management focuses on both neuroprotection and repair strategies, and is challenging due to low permeability of the blood-brain barrier, off-target distribution, and the need for high doses of drugs. The changes in the extracellular matrix have been documented in MS patients. It has been shown that the expression of nidogen-1 increases in MS lesions. Laminin forms a stable complex with nidogen-1 through a heptapeptide which was selected to target the lesion area in this study. Here we showed that the peptide binding was specific to the injured area following lysophosphatidylcholine (LPC) induced demyelination. In vivo data showed enhanced delivery of the peptide-functionalized gold nanoparticles (Pep-GNPs) to the lesion area. In addition, Pep-GNPs administration significantly enhanced myelin content and reduced astrocyte/microglia activation. Results demonstrated the possibility of using this peptide to target and treat lesions in patients suffering from MS.
{"title":"Peptide mediated targeted delivery of gold nanoparticles into the demyelination site ameliorates myelin impairment and gliosis","authors":"Sahar Farhangi MSc , Elham Karimi MSc , Khosro Khajeh PhD , Saman Hosseinkhani PhD , Mohammad Javan PhD","doi":"10.1016/j.nano.2022.102609","DOIUrl":"https://doi.org/10.1016/j.nano.2022.102609","url":null,"abstract":"<div><p>Drug development for multiple sclerosis (MS) clinical management focuses on both neuroprotection and repair strategies, and is challenging due to low permeability of the blood-brain barrier, off-target distribution, and the need for high doses of drugs. The changes in the extracellular matrix have been documented in MS patients. It has been shown that the expression of nidogen-1 increases in MS lesions. Laminin forms a stable complex with nidogen-1 through a heptapeptide which was selected to target the lesion area in this study. Here we showed that the peptide binding was specific to the injured area following lysophosphatidylcholine (LPC) induced demyelination. <em>In vivo</em> data showed enhanced delivery of the peptide-functionalized gold nanoparticles (Pep-GNPs) to the lesion area. In addition, Pep-GNPs administration significantly enhanced myelin content and reduced astrocyte/microglia activation. Results demonstrated the possibility of using this peptide to target and treat lesions in patients suffering from MS.</p></div>","PeriodicalId":396,"journal":{"name":"Nanomedicine: Nanotechnology, Biology and Medicine","volume":"47 ","pages":"Article 102609"},"PeriodicalIF":5.5,"publicationDate":"2023-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"3342725","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}