1
|
Mamnoon B, Moses AS, Sundaram S, Raitmayr CJ, Morgan T, Baldwin MK, Myatt L, Taratula O, Taratula OR. Glutathione-Responsive Methotrexate Polymersomes for Potential Management of Ectopic Pregnancy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2302969. [PMID: 37452511 PMCID: PMC10787806 DOI: 10.1002/smll.202302969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 06/22/2023] [Indexed: 07/18/2023]
Abstract
The first-line treatment for ectopic pregnancy (EP), the chemotherapeutic methotrexate (MTX), has a failure rate of more than 10%, which can lead to severe complications or death. Inadequate accumulation of administered MTX at the ectopic implantation site significantly contributes to therapeutic failure. This study reports the first glutathione-responsive polymersomes for efficient delivery of MTX to the implantation site and its triggered release in placental cells. Fluorescence and photoacoustic imaging have confirmed that the developed polymersomes preferentially accumulate after systemic administration in the implantation site of pregnant mice at early gestational stages. The high concentrations of intracellular glutathione (GSH) reduce an incorporated disulfide bond within polymersomes upon internalization into placental cells, resulting in their disintegration and efficient drug release. Consequently, MTX delivered by polymersomes induces pregnancy demise in mice, as opposed to free MTX at the same dose regimen. To achieve the same therapeutic efficacy with free MTX, a sixfold increase in dosage is required. In addition, mice successfully conceive and birth healthy pups following a prior complete pregnancy demise induced by methotrexate polymersomes. Therefore, the developed MTX nanomedicine can potentially improve EP management and reduce associated mortality rates and related cost.
Collapse
Affiliation(s)
- Babak Mamnoon
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
| | - Abraham S Moses
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
| | - Subisha Sundaram
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
| | - Constanze J Raitmayr
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
| | - Terry Morgan
- Department of Pathology and Laboratory Medicine, and the Center for Developmental Health, School of Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Maureen K Baldwin
- Department of Obstetrics and Gynecology, School of Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Leslie Myatt
- Department of Obstetrics and Gynecology, School of Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Oleh Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
| | - Olena R Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
| |
Collapse
|
2
|
Urbanova M, Cihova M, Buocikova V, Slopovsky J, Dubovan P, Pindak D, Tomas M, García-Bermejo L, Rodríguez-Garrote M, Earl J, Kohl Y, Kataki A, Dusinska M, Sainz B, Smolkova B, Gabelova A. Nanomedicine and epigenetics: New alliances to increase the odds in pancreatic cancer survival. Biomed Pharmacother 2023; 165:115179. [PMID: 37481927 DOI: 10.1016/j.biopha.2023.115179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/04/2023] [Accepted: 07/12/2023] [Indexed: 07/25/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is among the deadliest cancers worldwide, primarily due to its robust desmoplastic stroma and immunosuppressive tumor microenvironment (TME), which facilitate tumor progression and metastasis. In addition, fibrous tissue leads to sparse vasculature, high interstitial fluid pressure, and hypoxia, thereby hindering effective systemic drug delivery and immune cell infiltration. Thus, remodeling the TME to enhance tumor perfusion, increase drug retention, and reverse immunosuppression has become a key therapeutic strategy. In recent years, targeting epigenetic pathways has emerged as a promising approach to overcome tumor immunosuppression and cancer progression. Moreover, the progress in nanotechnology has provided new opportunities for enhancing the efficacy of conventional and epigenetic drugs. Nano-based drug delivery systems (NDDSs) offer several advantages, including improved drug pharmacokinetics, enhanced tumor penetration, and reduced systemic toxicity. Smart NDDSs enable precise targeting of stromal components and augment the effectiveness of immunotherapy through multiple drug delivery options. This review offers an overview of the latest nano-based approaches developed to achieve superior therapeutic efficacy and overcome drug resistance. We specifically focus on the TME and epigenetic-targeted therapies in the context of PDAC, discussing the advantages and limitations of current strategies while highlighting promising new developments. By emphasizing the immense potential of NDDSs in improving therapeutic outcomes in PDAC, our review paves the way for future research in this rapidly evolving field.
Collapse
Affiliation(s)
- Maria Urbanova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia
| | - Marina Cihova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia
| | - Verona Buocikova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia
| | - Jan Slopovsky
- 2nd Department of Oncology, National Cancer Institute, Klenova 1, 833 10 Bratislava, Slovakia; Faculty of Medicine, Comenius University, Spitalska 24, 813 72 Bratislava, Slovakia
| | - Peter Dubovan
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia; Department of Surgical Oncology, National CancerInstitute in Bratislava, Klenova 1, 833 10 Bratislava, Slovakia; Faculty of Medicine, Slovak Medical University in Bratislava, Limbová12, 833 03 Bratislava
| | - Daniel Pindak
- Department of Surgical Oncology, National CancerInstitute in Bratislava, Klenova 1, 833 10 Bratislava, Slovakia; Faculty of Medicine, Slovak Medical University in Bratislava, Limbová12, 833 03 Bratislava
| | - Miroslav Tomas
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia; Department of Surgical Oncology, National CancerInstitute in Bratislava, Klenova 1, 833 10 Bratislava, Slovakia; Faculty of Medicine, Slovak Medical University in Bratislava, Limbová12, 833 03 Bratislava
| | - Laura García-Bermejo
- Biomarkers and Therapeutic Targets Group, Area4, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9100, 28034 Madrid, Spain
| | - Mercedes Rodríguez-Garrote
- Molecular Epidemiology and Predictive Tumor Markers Group, Area 3, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9100, 28034 Madrid, Spain; CIBERONC, Madrid, Spain
| | - Julie Earl
- Molecular Epidemiology and Predictive Tumor Markers Group, Area 3, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9100, 28034 Madrid, Spain; CIBERONC, Madrid, Spain
| | - Yvonne Kohl
- Department Bioprocessing & Bioanalytics, Fraunhofer Institute for Biomedical Engineering IBMT, 66280 Sulzbach, Germany
| | - Agapi Kataki
- 1st Department of Propaedeutic Surgery, National and Kapodistrian University of Athens, Vasilissis Sofias 114, 11527 Athens, Greece
| | - Maria Dusinska
- Health Effects Laboratory, Department of Environmental Chemistry, NILU-Norwegian Institute for Air Research, Instituttveien 18, 2002 Kjeller, Norway
| | - Bruno Sainz
- CIBERONC, Madrid, Spain; Instituto de Investigaciones Biomédicas"Alberto Sols" (IIBM), CSIC-UAM, 28029 Madrid, Spain; Biomarkers and Personalized Approach to Cancer (BIOPAC) Group, Area 3, Ramón y Cajal Health Research Institute (IRYCIS), 28034 Madrid, Spain
| | - Bozena Smolkova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia
| | - Alena Gabelova
- Department of Nanobiology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 84505 Bratislava, Slovakia..
| |
Collapse
|
3
|
Zhang H, Cao K, Xiang J, Zhang M, Zhu M, Xi Q. Hypoxia induces immunosuppression, metastasis and drug resistance in pancreatic cancers. Cancer Lett 2023; 571:216345. [PMID: 37558084 DOI: 10.1016/j.canlet.2023.216345] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/26/2023] [Accepted: 08/04/2023] [Indexed: 08/11/2023]
Abstract
Pancreatic cancer is one of the common malignant tumors of the digestive system and is known as the "king of cancers". It is extremely difficult to diagnose at an early stage, the disease progresses rapidly, and the effect of chemotherapy and radiotherapy is poor, so the prognosis of pancreatic cancer patients is very poor. Numerous studies have suggested that hypoxia is closely related to the development and progression of pancreatic cancer. Inadequate blood supply and desmoplasia in the microenvironment of pancreatic cancer can result in its extreme hypoxia. This hypoxic microenvironment can further contribute to angiogenesis and desmoplasia. Hypoxia is mediated by the complex hypoxia inducible factor (HIF) signaling pathway and plays an important role in the formation of a highly immunosuppressive microenvironment and the metastasis of pancreatic cancer. Further work on the hypoxic microenvironment will help clarify the specific mechanisms of the role of hypoxia in pancreatic cancer and provide a basis for the realization of hypoxia-targeted therapeutic and diagnostic strategies.
Collapse
Affiliation(s)
- Huan Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Kailei Cao
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Jingrong Xiang
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Mengting Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Mengxin Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Qinhua Xi
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
4
|
Ren M, Feng L, Zong R, Sun H. Novel prognostic gene signature for pancreatic ductal adenocarcinoma based on hypoxia. World J Surg Oncol 2023; 21:257. [PMID: 37605192 PMCID: PMC10464224 DOI: 10.1186/s12957-023-03142-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/08/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND Currently, there is lack of marker to accurately assess the prognosis of patients diagnosed with pancreatic ductal adenocarcinoma (PDAC). This study aims to establish a hypoxia-related risk scoring model that can effectively predict the prognosis and chemotherapy outcomes of PDAC patients. METHODS Using unsupervised consensus clustering algorithms, we comprehensively analyzed The Cancer Genome Atlas (TCGA) data to identify two distinct hypoxia clusters and used the weighted gene co-expression network analysis (WGCNA) to examine gene sets significantly associated with these hypoxia clusters. Then univariate Cox regression, the least absolute shrinkage and selection operator (LASSO) Cox regression and multivariate Cox regression were used to construct a signature and its efficacy was evaluated using the International Cancer Genome Consortium (ICGC) PDAC cohort. Further, the correlation between the risk scores obtained from the signature and carious clinical, pathological, immunophenotype, and immunoinfiltration factors as well as the differences in immunotherapy potential and response to common chemotherapy drugs between high-risk and low-risk groups were evaluated. RESULTS From a total of 8 significantly related modules and 4423 genes, 5 hypoxia-related signature genes were identified to construct a risk model. Further analysis revealed that the overall survival rate (OS) of patients in the low-risk group was significantly higher than the high-risk group. Univariate and multivariate Cox regression analysis showed that the risk scoring signature was an independent factor for prognosis prediction. Analysis of immunocyte infiltration and immunophenotype showed that the immune score and the anticancer immune response in the high-risk were significantly lower than that in the low-risk group. CONCLUSION The constructed hypoxia-associated prognostic signature demonstrated could be used as a potential risk classifier for PDAC.
Collapse
Affiliation(s)
- Min Ren
- College of Life Science, Yan'an University, Yan'an, 716000, China
| | - Liaoliao Feng
- College of Life Science, Yan'an University, Yan'an, 716000, China
| | - Rongrong Zong
- College of Life Science, Yan'an University, Yan'an, 716000, China
| | - Huiru Sun
- College of Life Science, Yan'an University, Yan'an, 716000, China.
| |
Collapse
|
5
|
Zlotnikov ID, Ezhov AA, Ferberg AS, Krylov SS, Semenova MN, Semenov VV, Kudryashova EV. Polymeric Micelles Formulation of Combretastatin Derivatives with Enhanced Solubility, Cytostatic Activity and Selectivity against Cancer Cells. Pharmaceutics 2023; 15:1613. [PMID: 37376064 DOI: 10.3390/pharmaceutics15061613] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/26/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Combretastatin derivatives is a promising class of antitumor agents, tubulin assembly inhibitors. However, due to poor solubility and insufficient selectivity to tumor cells, we believe, their therapeutic potential has not been fully realized yet. This paper describes polymeric micelles based on chitosan (a polycation that causes pH and thermosensitivity of micelles) and fatty acids (stearic, lipoic, oleic and mercaptoundecanoic), which were used as a carrier for a range of combretastatin derivatives and reference organic compounds, demonstrating otherwise impossible delivery to tumor cells, at the same time substantially reduced penetration into normal cells. Polymers containing sulfur atoms in hydrophobic tails form micelles with a zeta potential of about 30 mV, which increases to 40-45 mV when cytostatics are loaded. Polymers with tails of oleic and stearic acids form poorly charged micelles. The use of polymeric 400 nm micelles provides the dissolution of hydrophobic potential drug molecules. Micelles could significantly increase the selectivity of cytostatics against tumors, which has been shown using MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, Fourier transform infrared (FTIR) spectroscopy, flow cytometry and fluorescence microscopy. Atomic force microscopy presented the difference between the unloaded micelles and those loaded with the drug: the size of the former was 30 nm on average, while the latter had a "disc-like" shape and a size of about 450 nm. The loading of drugs into the core of micelles was confirmed by UV and fluorescence spectroscopy methods; shifts of absorption and emission maxima into the long-wavelength region by tens of nm was observed. With FTIR spectroscopy, a high interaction efficiency of micelles with the drug on cells was demonstrated, but at the same time, selective absorption was observed: micellar cytostatics penetrate into A549 cancer cells 1.5-2 times better than the simple form of the drugs. Moreover, in normal HEK293T, the penetration of the drug is reduced. The proposed mechanism for reducing the accumulation of drugs in normal cells is the adsorption of micelles on the cell surface and the preservation of cytostatics to penetrate inside the cells. At the same time, in cancer cells, due to the structural features of the micelles, they penetrate inside, merging with the membrane and releasing the drug by pH- and glutathione-sensitive mechanisms. From a methodological point of view, we have proposed a powerful approach to the observation of micelles using a flow cytometer, which, in addition, allows us to quantify the cells that have absorbed/adsorbed cytostatic fluorophore and distinguish between specific and non-specific binding. Thus, we present polymeric micelles as drug delivery systems in tumors using the example of combretastatin derivatives and model fluorophore-cytostatic rhodamine 6G.
Collapse
Affiliation(s)
- Igor D Zlotnikov
- Faculty of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia
| | - Alexander A Ezhov
- Faculty of Physics, Lomonosov Moscow State University, Leninskie Gory 1/2, 119991 Moscow, Russia
| | - Artem S Ferberg
- Faculty of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia
| | - Sergey S Krylov
- N. D. Zelinsky Institute of Organic Chemistry RAS, Leninsky Prospect 47, 119991 Moscow, Russia
| | - Marina N Semenova
- N. K. Koltzov Institute of Developmental Biology RAS, Vavilov Street 26, 119334 Moscow, Russia
| | - Victor V Semenov
- N. D. Zelinsky Institute of Organic Chemistry RAS, Leninsky Prospect 47, 119991 Moscow, Russia
| | - Elena V Kudryashova
- Faculty of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia
| |
Collapse
|
6
|
Boria AJ, Narayanasamy G, Bimali M, Maraboyina S, Kalantari F, Sabouri P, Su Z. Cleaning the dose falloff with low modulation in SBRT lung plans. Biomed Phys Eng Express 2023; 9. [PMID: 37140156 DOI: 10.1088/2057-1976/acd008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/25/2023] [Indexed: 05/05/2023]
Abstract
Purpose.This dosimetric study is intended to lower the modulation factor in lung SBRT plans generated in the Eclipse TPS that could replace highly modulated plans that are prone to the interplay effect.Materials and methods.Twenty clinical lung SBRT plans with high modulation factors (≥4) were replanned in Varian Eclipse TPS version 15.5 utilizing 2 mm craniocaudal and 1 mm axial block margins followed by light optimization in order to reduce modulation. A unique plan optimization methodology, which utilizes a novel shell structure (OptiForR50) for R50%optimization in addition to five consecutive concentric 5 mm shells, was utilized to control dose falloff according to RTOG 0813 and 0915 recommendations. The prescription varied from 34-54 Gy in 1-4 fractions, and the dose objectives were PTV D95%= Rx, PTV Dmax< 140% of Rx, and minimizing the modulation factor. Plan evaluation metrics included modulation factor, CIRTOG, homogeneity index (HI), R50%, D2cm, V105%, and lung V8-12.8Gy(Timmerman Constraint). A random-intercept linear mixed effects model was used with a p ≤ 0.05 threshold to test for statistical significance.Results.The retrospectively generated plans had significantly lower modulation factors (3.65 ± 0.35 versus 4.59 ± 0.54; p < 0.001), lower CIRTOG(0.97 ± 0.02 versus 1.02 ± 0.06; p = 0.001), higher HI (1.35 ± 0.06 versus 1.14 ± 0.04; p < 0.001), lower R50%(4.09 ± 0.45 versus 4.56 ± 0.56; p < 0.001), and lower lungs V8-12.8Gy(Timmerman) (4.61% ± 3.18% versus 4.92% ± 3.37%; p < 0.001). The high dose spillage V105%was borderline significantly lower (0.44% ± 0.49% versus 1.10% ± 1.64%; p = 0.051). The D2cmwas not statistically different (46.06% ± 4.01% versus 46.19% ± 2.80%; p = 0.835).Conclusion.Lung SBRT plans with significantly lower modulation factors can be generated that meet the RTOG constraints, using our planning strategy.
Collapse
Affiliation(s)
- Andrew J Boria
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, AR, United States of America
| | - Ganesh Narayanasamy
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, AR, United States of America
| | - Milan Bimali
- Nexus Institute for Research and Innovation, Lalitpur, Nepal
| | - Sanjay Maraboyina
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, AR, United States of America
| | - Faraz Kalantari
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, AR, United States of America
| | - Pouya Sabouri
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, AR, United States of America
| | - Zhong Su
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, AR, United States of America
| |
Collapse
|
7
|
Suneetha M, Kim H, Han SS. Doxorubicin-Loaded Fungal-Carboxymethyl Chitosan Functionalized Polydopamine Nanoparticles for Photothermal Cancer Therapy. Pharmaceutics 2023; 15:pharmaceutics15041281. [PMID: 37111766 PMCID: PMC10142010 DOI: 10.3390/pharmaceutics15041281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 04/11/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
In this work, we synthesized doxorubicin-loaded fungal-carboxymethyl chitosan (FC) functionalized polydopamine (Dox@FCPDA) nanoparticles for improved anticancer activity via photothermal drug release. The photothermal properties revealed that the FCPDA nanoparticles with a concentration of 400 µg/mL produced a temperature of about 61.1 °C at 2 W/cm2 laser illumination, which is more beneficial for cancer cells. Due to the hydrophilic FC biopolymer, the Dox was successfully encapsulated into FCPDA nanoparticles via electrostatic interactions and pi-pi stacking. The maximum drug loading and encapsulation efficiency were calculated to be 19.3% and 80.2%, respectively. The Dox@FCPDA nanoparticles exhibited improved anticancer activity on HePG2 cancer cells when exposed to an NIR laser (800 nm, 2 W/cm2). Furthermore, the Dox@FCPDA nanoparticles also improved cellular uptake with HepG2 cells. Therefore, functionalizing FC biopolymer with PDA nanoparticles is more beneficial for drug and photothermal dual therapeutic properties for cancer therapy.
Collapse
Affiliation(s)
- Maduru Suneetha
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Hyeonjin Kim
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
8
|
Li X, Yang X, Xue W, Yang R, He Z, Ai L, Liu H. Identification of gene signatures related to hypoxia and angiogenesis in pancreatic cancer to aid immunotherapy and prognosis. Front Oncol 2023; 13:1119763. [PMID: 37064125 PMCID: PMC10098147 DOI: 10.3389/fonc.2023.1119763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 03/10/2023] [Indexed: 04/03/2023] Open
Abstract
BackgroundOne of the most diverse tumors is pancreatic cancer (PC), which makes predicting the prognosis challenging. PC development is directly related to hypoxia, angiogenesis, and immunotherapy. It is still unclear how the three features are related.MethodsThe Genotype-Tissue Expression (GTEx) and the Cancer Genome Atlas (TCGA) database were employed to obtain sequencing data for healthy pancreatic tissues and PC tissues, respectively. According to the constructed hypoxic prognostic model (HPM) and angiogenic prognostic model (APM), 4 subtypes of PC were identified. Hypoxia and angiogenesis prognostic model (HAPM) was established based on differentially expressed genes (DEGs) between high-angiogenesis/high-hypoxia (HH) and low-angiogenesis/low-hypoxia (LL) subgroups. Base on the median risk score, PC patients were separated into high-risk and low-risk groups, and clinical traits, prognosis, percentage of immune cell infiltration, PD-1 expression, and the fraction of T-cell depletion were compared between the groups. Finally, the predictive accuracy of the tumor immune dysfunction and rejection (TIDE) and tumor inflammatory signature (TIS) models, as well as HAPM, was compared.ResultWe analyzed the mRNA sequencing data from 178 PC tissues and 171 normal pancreatic tissues to obtain 9527 DEGs. We discovered 200 genes linked with hypoxia and 36 genes involved with angiogenesis through the literature. We found the core genes related with hypoxia and angiogenesis in PC by intersecting the DEGs of the HH and LL subgroups with those of PC via WGCNA. IL-17 signaling pathway, ECM-receptor interactions, cytokine receptor interactions, etc. were all enriched in the Kyoto Encyclopedia of Genes and Genomes (KEGG) results of core genes. HAPM has good predictive efficiency, according to an evaluation of KM survival curves and ROC curves. The external dataset also validated the model’s ability to anticipate outcomes. Patients in the high- and low-risk groups were compared for PD1 expression and T-cell exclusion scores, which suggested that the model might be used to forecast which PC patients might benefit from immunotherapy.ConclusionsThe probable molecular processes connecting hypoxia and angiogenesis are described in this work, and a model is developed that may be utilized to forecast the prognosis for PC patients and the benefits of immunotherapy.
Collapse
Affiliation(s)
- Xiushen Li
- Department of Obstetrics and Gynaecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Xi Yang
- Department of Ultrasound, The People’s Hospital of Shapingba District, Chongqing, China
| | - Weiqi Xue
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Rui Yang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, Guangdong, China
- Department of Hepatobiliary Surgery, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
| | - Zhiwei He
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, Guangdong, China
- Department of Hepatobiliary Surgery, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
| | - Lisha Ai
- Department of Teaching and Research, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
| | - Hui Liu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, Guangdong, China
- Department of Hepatobiliary Surgery, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases and Carson International Cancer, Shenzhen University, Shenzhen, China
- *Correspondence: Hui Liu,
| |
Collapse
|
9
|
Hughes KA, Misra B, Maghareh M, Bobbala S. Use of stimulatory responsive soft nanoparticles for intracellular drug delivery. NANO RESEARCH 2023; 16:6974-6990. [PMID: 36685637 PMCID: PMC9840428 DOI: 10.1007/s12274-022-5267-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/30/2022] [Accepted: 10/31/2022] [Indexed: 05/24/2023]
Abstract
Drug delivery has made tremendous advances in the last decade. Targeted therapies are increasingly common, with intracellular delivery highly impactful and sought after. Intracellular drug delivery systems have limitations due to imprecise and non-targeted release profiles. One way this can be addressed is through using stimuli-responsive soft nanoparticles, which contain materials with an organic backbone such as lipids and polymers. The choice of biomaterial is essential for soft nanoparticles to be responsive to internal or external stimuli. The nanoparticle must retain its integrity and payload in non-targeted physiological conditions while responding to particular intracellular environments where payload release is desired. Multiple internal and external factors could stimulate the intracellular release of drugs from nanoparticles. Internal stimuli include pH, oxidation, and enzymes, while external stimuli include ultrasound, light, electricity, and magnetic fields. Stimulatory responsive soft nanoparticulate systems specifically utilized to modulate intracellular delivery of drugs are explored in this review.
Collapse
Affiliation(s)
- Krystal A. Hughes
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV 26505 USA
| | - Bishal Misra
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV 26505 USA
| | - Maryam Maghareh
- Department of Clinical Pharmacy, West Virginia University School of Pharmacy, Morgantown, WV 26505 USA
| | - Sharan Bobbala
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV 26505 USA
| |
Collapse
|
10
|
Prospects for hypoxia-based drug delivery platforms for the elimination of advanced metastatic tumors: From 3D modeling to clinical concepts. J Control Release 2023; 353:1002-1022. [PMID: 36516901 DOI: 10.1016/j.jconrel.2022.12.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 11/30/2022] [Accepted: 12/03/2022] [Indexed: 12/24/2022]
Abstract
Hypoxia is a unique characteristic of the solid tumor microenvironment. Hypoxia contributes to multi-drug resistance, metastasis and cancer relapse through numerous molecular pathways, but at the same time provides an opportunity for the development of novel drugs or modalities specifically targeting hypoxic tumor regions. Given the high significance of tumor hypoxia in therapeutic results, we here discuss a variety of hypoxia-adopted strategies, and their potential and utility in the treatment of deep-seated hypoxic tumor cells. We discuss the merits and demerits of these approaches, as well as their combination with other approaches such as photodynamic therapy. We also survey the currently available 3D hypoxia modeling systems, in particular organoid-based microfluidics. Finally, we discuss the potential and the current status of preclinical tumor hypoxia approaches in clinical trials for advanced cancer. We believe that multi-modal imaging and therapeutic hypoxia adopted drug delivery platforms could provide better efficacy and safety profiles, and more importantly personalized therapy. Determining the hypoxia status of tumors could offer a second chance for the clinical translation of hypoxia-based agents, such as hypoxia activated prodrugs (HAPs) from bench to bedside.
Collapse
|
11
|
Swetha KL, Maravajjala KS, Li SD, Singh MS, Roy A. Breaking the niche: multidimensional nanotherapeutics for tumor microenvironment modulation. Drug Deliv Transl Res 2023; 13:105-134. [PMID: 35697894 DOI: 10.1007/s13346-022-01194-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2022] [Indexed: 12/13/2022]
Abstract
Most of the current antitumor therapeutics were developed targeting the cancer cells only. Unfortunately, in the majority of tumors, this single-dimensional therapy is found to be ineffective. Advanced research has shown that cancer is a multicellular disorder. The tumor microenvironment (TME), which is made by a complex network of the bulk tumor cells and other supporting cells, plays a crucial role in tumor progression. Understanding the importance of the TME in tumor growth, different treatment modalities have been developed targeting these supporting cells. Recent clinical results suggest that simultaneously targeting multiple components of the tumor ecosystem with drug combinations can be highly effective. This type of "multidimensional" therapy has a high potential for cancer treatment. However, tumor-specific delivery of such multi-drug combinations remains a challenge. Nanomedicine could be utilized for the tumor-targeted delivery of such multidimensional therapeutics. In this review, we first give a brief overview of the major components of TME. We then highlight the latest developments in nanoparticle-based combination therapies, where one drug targets cancer cells and other drug targets tumor-supporting components in the TME for a synergistic effect. We include the latest preclinical and clinical studies and discuss innovative nanoparticle-mediated targeting strategies.
Collapse
Affiliation(s)
- K Laxmi Swetha
- Department of Pharmacy, Birla Institute of Technology & Science, Vidya Vihar, Pilani, Rajasthan, 333031, India
| | - Kavya Sree Maravajjala
- Department of Pharmacy, Birla Institute of Technology & Science, Vidya Vihar, Pilani, Rajasthan, 333031, India
| | - Shyh-Dar Li
- Faculty of Pharmaceutical Sciences, The University of British Columbia, 2405 Westbrook Mall, Vancouver, BC, Canada
| | - Manu Smriti Singh
- Department of Biotechnology, Bennett University, Greater Noida, Uttar Pradesh, 201310, India. .,Center of Excellence for Nanosensors and Nanomedicine, Bennett University, Greater Noida, Uttar Pradesh, 201310, India.
| | - Aniruddha Roy
- Department of Pharmacy, Birla Institute of Technology & Science, Vidya Vihar, Pilani, Rajasthan, 333031, India.
| |
Collapse
|
12
|
Sim TM. Nanoparticle-assisted targeting of the tumour microenvironment. OPENNANO 2022. [DOI: 10.1016/j.onano.2022.100097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
13
|
Busato D, Mossenta M, Dal Bo M, Macor P, Toffoli G. The Proteoglycan Glypican-1 as a Possible Candidate for Innovative Targeted Therapeutic Strategies for Pancreatic Ductal Adenocarcinoma. Int J Mol Sci 2022; 23:ijms231810279. [PMID: 36142190 PMCID: PMC9499405 DOI: 10.3390/ijms231810279] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/02/2022] [Accepted: 09/04/2022] [Indexed: 11/19/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) accounts for 90% of all pancreatic cancers, with a 5-year survival rate of 7% and 80% of patients diagnosed with advanced or metastatic malignancies. Despite recent advances in diagnostic testing, surgical techniques, and systemic therapies, there remain limited options for the effective treatment of PDAC. There is an urgent need to develop targeted therapies that are able to differentiate between cancerous and non-cancerous cells to reduce side effects and better inhibit tumor growth. Antibody-targeted strategies are a potentially effective option for introducing innovative therapies. Antibody-based immunotherapies and antibody-conjugated nanoparticle-based targeted therapies with antibodies targeting specific tumor-associated antigens (TAA) can be proposed. In this context, glypican-1 (GPC1), which is highly expressed in PDAC and not expressed or expressed at very low levels in non-malignant lesions and healthy pancreatic tissues, is a useful TAA that can be achieved by a specific antibody-based immunotherapy and antibody-conjugated nanoparticle-based targeted therapy. In this review, we describe the main clinical features of PDAC. We propose the proteoglycan GPC1 as a useful TAA for PDAC-targeted therapies. We also provide a digression on the main developed approaches of antibody-based immunotherapy and antibody-conjugated nanoparticle-based targeted therapy, which can be used to target GPC1.
Collapse
Affiliation(s)
- Davide Busato
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
- Correspondence: ; Tel.: +39-0434-659816
| | - Monica Mossenta
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| | - Paolo Macor
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| |
Collapse
|
14
|
Khakinahad Y, Sohrabi S, Razi S, Narmani A, Khaleghi S, Asadiyun M, Jafari H, Mohammadnejad J. Margetuximab conjugated-PEG-PAMAM G4 nano-complex: a smart nano-device for suppression of breast cancer. Biomed Eng Lett 2022; 12:317-329. [PMID: 35892030 PMCID: PMC9308845 DOI: 10.1007/s13534-022-00225-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 01/06/2022] [Accepted: 03/06/2022] [Indexed: 12/18/2022] Open
Abstract
Abstract Breast cancer due to its high incidence and mortality is the second leading cause of death among females. On the other hand, nanoparticle-based drug delivery is one of the most promising approaches in cancer therapy, nowadays. Hence, margetuximab- and polyethylene glycol-conjugated PAMAM G4 dendrimers were efficiently synthesized for targeted delivery of quercetin (therapeutic agent) to MDA-MB-231 breast cancer cells. Synthesized nano-complexes were characterized using analytical devices such as FT-IR, TGA, DLS, Zeta potential analyzer, and TEM. The size less than 40 nm, - 18.8 mV surface charge, efficient drug loading capacity (21.48%), and controlled drug release (about 45% of drug release normal pH after 8 h) were determined for the nano-complex. In the biomedical test, the cell viability was obtained 14.67% at 24 h of post-treatment for 800 nM concentration, and IC50 was ascertained at 100 nM for the nano-complex. The expression of apoptotic Bax and Caspase9 genes was increased by more than eightfolds and more than fivefolds after treatment with an optimal concentration of nanocarrier. Also, more than threefolds of cell cycle arrest was observed at the optimal concentration synthetics, and 27.5% breast cancer cell apoptosis was detected after treatment with 100 nM nano-complex. These outputs have been indicating the potential capacity of synthesized nano-complex in inhibiting the growth of breast cancer cells. Graphic abstract
Collapse
Affiliation(s)
- Yasaman Khakinahad
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
- Department of Biological and Biomedical Sciences, Cancer Biomedical Center, Tehran, Iran
| | - Saeedeh Sohrabi
- Department of Biological and Biomedical Sciences, Cancer Biomedical Center, Tehran, Iran
- Department of Biology, Faculty of Advanced Sciences and Technology, Payam Noor University, Tehran, Iran
| | - Shokufeh Razi
- Department of Biological and Biomedical Sciences, Cancer Biomedical Center, Tehran, Iran
- Department of Genetics, Faculty of Basic Sciences, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Asghar Narmani
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Sepideh Khaleghi
- Department of Medical Biotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahboubeh Asadiyun
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
- Department of Biological and Biomedical Sciences, Cancer Biomedical Center, Tehran, Iran
| | - Hanieh Jafari
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Javad Mohammadnejad
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| |
Collapse
|
15
|
Sarkar D, Chowdhury M, Das PK. Naphthalimide-Based Azo-Functionalized Supramolecular Vesicle in Hypoxia-Responsive Drug Delivery. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:3480-3492. [PMID: 35261245 DOI: 10.1021/acs.langmuir.1c03334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Supramolecular materials that respond to external triggers are being extensively utilized in developing spatiotemporal control in biomedical applications ranging from drug delivery to diagnostics. The present article describes the development of self-assembled vesicles in 1:9 (v/v), tetrahydrofuran (THF)-water by naphthalimide-based azo moiety containing amphiphile (NI-Azo) where azo moiety would act as the stimuli-responsive junction. The self-assembly of NI-Azo took place through H-type of aggregation. Microscopic and spectroscopic analyses confirmed the formation of supramolecular vesicles with a dimension of 200-250 nm. Azo (-N═N-) moiety is known to get reduced to amine derivatives in the presence of the azoreductase enzyme, which is overexpressed in the hypoxic microenvironment. The absorbance intensity of this characteristic azo (-N═N-) moiety of NI-Azo (1:9 (v/v), THF-water) at 458 nm got diminished in the presence of both extracellular and intracellular bacterial azoreductase extracted from Escherichia coli bacteria. The same observation was noted in the presence of sodium dithionite (mimic of azoreductase), indicating that azoreductase/sodium dithionite induced azo bond cleavage of NI-Azo, which was confirmed by matrix-assisted laser desorption ionization time-of-flight spectrometric data of the corresponding aromatic amine fragments. The anticancer drug, curcumin, was encapsulated inside NI-Azo vesicles that successfully killed B16F10 cells (cancer cells) in CoCl2-induced hypoxic environment owing to the azoreductase-responsive release of drug. The cancer cell killing efficiency by curcumin-loaded NI-Azo vesicles in the hypoxic condition was 2.15-fold higher than that of the normoxic environment and 2.4-fold higher compared to that of native curcumin in the hypoxic condition. Notably, cancer cell killing efficiency of curcumin-loaded NI-Azo vesicles was 4.5- and 1.9-fold higher than that of noncancerous NIH3T3 cells in normoxic and hypoxic environments, respectively. Cell killing was found to be primarily through the early apoptotic pathway.
Collapse
Affiliation(s)
- Deblina Sarkar
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Monalisa Chowdhury
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Prasanta Kumar Das
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| |
Collapse
|
16
|
Alhalhooly L, Confeld MI, Woo SO, Mamnoon B, Jacobson R, Ghosh S, Kim J, Mallik S, Choi Y. Single-Molecule Force Probing of RGD-Binding Integrins on Pancreatic Cancer Cells. ACS APPLIED MATERIALS & INTERFACES 2022; 14:7671-7679. [PMID: 35113515 PMCID: PMC8890904 DOI: 10.1021/acsami.1c23361] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Integrin-targeting arginine-glycine-aspartic acid (RGD)-based nanocarriers have been widely used for tumor imaging, monitoring of tumor development, and delivery of anticancer drugs. However, the thermodynamics of an RGD-integrin formation and dissociation associated with binding dynamics, affinity, and stability remains unclear. Here, we probed the binding strength of the binary complex to live pancreatic cancer cells using single-molecule binding force spectroscopy methods, in which RGD peptides were functionalized on a force probe tip through poly(ethylene glycol) (PEG)-based bifunctional linker molecules. While the density of integrin αV receptors on the cell surface varies more than twofold from cell line to cell line, the individual RGD-integrin complexes exhibited a cell type-independent, monovalent bond strength. The load-dependent bond strength of multivalent RGD-integrin interactions scaled sublinearly with increasing bond number, consistent with the noncooperative, parallel bond model. Furthermore, the multivalent bonds ruptured sequentially either by one or in multiples, and the force strength was comparable to the synchronous rupture force. Comparison of energy landscapes of the bond number revealed a substantial decrease of kinetic off-rates for multivalent bonds, along with the increased width of the potential well and the increased potential barrier height between bound and unbound states, enhancing the stability of the multivalent bonds between them.
Collapse
Affiliation(s)
- Lina Alhalhooly
- Department of Physics, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Matthew I. Confeld
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Sung Oh Woo
- Department of Physics, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Babak Mamnoon
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Reed Jacobson
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Shrinwanti Ghosh
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Jiha Kim
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota 58108, United State
- Molecular and Cellular Biology Program, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Sanku Mallik
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Yongki Choi
- Department of Physics, North Dakota State University, Fargo, North Dakota 58108, United State
- Molecular and Cellular Biology Program, North Dakota State University, Fargo, North Dakota 58108, United State
- Materials and Nanotechnology Program, North Dakota State University, Fargo, North Dakota 58108, United State
| |
Collapse
|
17
|
Nanomedicine in Pancreatic Cancer: Current Status and Future Opportunities for Overcoming Therapy Resistance. Cancers (Basel) 2021; 13:cancers13246175. [PMID: 34944794 PMCID: PMC8699181 DOI: 10.3390/cancers13246175] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/12/2021] [Accepted: 11/16/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Despite access to a vast arsenal of anticancer agents, many fail to realise their full therapeutic potential in clinical practice. One key determinant of this is the evolution of multifaceted resistance mechanisms within the tumour that may either pre-exist or develop during the course of therapy. This is particularly evident in pancreatic cancer, where limited responses to treatment underlie dismal survival rates, highlighting the urgent need for new therapeutic approaches. Here, we discuss the major features of pancreatic tumours that contribute to therapy resistance, and how they may be alleviated through exploitation of the mounting and exciting promise of nanomedicines; a unique collection of nanoscale platforms with tunable and multifunctional capabilities that have already elicited a widespread impact on cancer management. Abstract The development of drug resistance remains one of the greatest clinical oncology challenges that can radically dampen the prospect of achieving complete and durable tumour control. Efforts to mitigate drug resistance are therefore of utmost importance, and nanotechnology is rapidly emerging for its potential to overcome such issues. Studies have showcased the ability of nanomedicines to bypass drug efflux pumps, counteract immune suppression, serve as radioenhancers, correct metabolic disturbances and elicit numerous other effects that collectively alleviate various mechanisms of tumour resistance. Much of this progress can be attributed to the remarkable benefits that nanoparticles offer as drug delivery vehicles, such as improvements in pharmacokinetics, protection against degradation and spatiotemporally controlled release kinetics. These attributes provide scope for precision targeting of drugs to tumours that can enhance sensitivity to treatment and have formed the basis for the successful clinical translation of multiple nanoformulations to date. In this review, we focus on the longstanding reputation of pancreatic cancer as one of the most difficult-to-treat malignancies where resistance plays a dominant role in therapy failure. We outline the mechanisms that contribute to the treatment-refractory nature of these tumours, and how they may be effectively addressed by harnessing the unique capabilities of nanomedicines. Moreover, we include a brief perspective on the likely future direction of nanotechnology in pancreatic cancer, discussing how efforts to develop multidrug formulations will guide the field further towards a therapeutic solution for these highly intractable tumours.
Collapse
|
18
|
Mortezaee K, Majidpoor J. The impact of hypoxia on immune state in cancer. Life Sci 2021; 286:120057. [PMID: 34662552 DOI: 10.1016/j.lfs.2021.120057] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/10/2021] [Accepted: 10/12/2021] [Indexed: 12/14/2022]
Abstract
Hypoxia is a known feature of solid tumors and a critical promoter of tumor hallmarks. Hypoxia influences tumor immunity in a way favoring immune evasion and resistance. Extreme hypoxia and aberrant hypoxia-inducible factor-1 (HIF-1) activity in tumor microenvironment (TME) is a drawback for effective immunotherapy. Infiltration and activity of CD8+ T cells is reduced in such condition, whereas regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) show high activities. Highly hypoxic TME also impairs maturation and activity of dendritic cell (DCs) and natural killer (NK) cells. In addition, the hypoxic TME positively is linked positively with metabolic changes in cells of immune system. These alterations are indicative of a need for hypoxia modulation as a complementary targeting strategy to go with immune checkpoint inhibitor (ICI) therapy.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Disease Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
19
|
Jia M, Zhang D, Zhang C, Li C. Nanoparticle-based delivery systems modulate the tumor microenvironment in pancreatic cancer for enhanced therapy. J Nanobiotechnology 2021; 19:384. [PMID: 34809634 PMCID: PMC8607729 DOI: 10.1186/s12951-021-01134-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/12/2021] [Indexed: 02/08/2023] Open
Abstract
Pancreatic cancer is one of the most lethal malignant tumors with a low survival rate, partly because the tumor microenvironment (TME), which consists of extracellular matrix (ECM), cancer-associated fibroblasts (CAFs), immune cells, and vascular systems, prevents effective drug delivery and chemoradiotherapy. Thus, modulating the microenvironment of pancreatic cancer is considered a promising therapeutic approach. Since nanoparticles are one of the most effective cancer treatment strategies, several nano-delivery platforms have been developed to regulate the TME and enhance treatment. Here, we summarize the latest advances in nano-delivery systems that alter the TME in pancreatic cancer by depleting ECM, inhibiting CAFs, reversing immunosuppression, promoting angiogenesis, or improving the hypoxic environment. We also discuss promising new targets for such systems. This review is expected to improve our understanding of how to modulate the pancreatic cancer microenvironment and guide the development of new therapies.
Collapse
Affiliation(s)
- Ming Jia
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, No.1, Section 1, Xianglin Road, Luzhou, Sichuan, 646000, People's Republic of China
| | - Dan Zhang
- Department of Pharmacy of Traditional Chinese Medicine, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Chunxiang Zhang
- The Key Laboratory of Medical Electrophysiology of the Ministry of Education, Southwest Medical University, No.1, Section 1, Xianglin Road, Luzhou, Sichuan, 646000, People's Republic of China.
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, No.1, Section 1, Xianglin Road, Luzhou, Sichuan, 646000, People's Republic of China.
| |
Collapse
|
20
|
The Evolution and Future of Targeted Cancer Therapy: From Nanoparticles, Oncolytic Viruses, and Oncolytic Bacteria to the Treatment of Solid Tumors. NANOMATERIALS 2021; 11:nano11113018. [PMID: 34835785 PMCID: PMC8623458 DOI: 10.3390/nano11113018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/28/2021] [Accepted: 11/01/2021] [Indexed: 02/07/2023]
Abstract
While many classes of chemotherapeutic agents exist to treat solid tumors, few can generate a lasting response without substantial off-target toxicity despite significant scientific advancements and investments. In this review, the paths of development for nanoparticles, oncolytic viruses, and oncolytic bacteria over the last 20 years of research towards clinical translation and acceptance as novel cancer therapeutics are compared. Novel nanoparticle, oncolytic virus, and oncolytic bacteria therapies all start with a common goal of accomplishing therapeutic drug activity or delivery to a specific site while avoiding off-target effects, with overlapping methodology between all three modalities. Indeed, the degree of overlap is substantial enough that breakthroughs in one therapeutic could have considerable implications on the progression of the other two. Each oncotherapeutic modality has accomplished clinical translation, successfully overcoming the potential pitfalls promising therapeutics face. However, once studies enter clinical trials, the data all but disappears, leaving pre-clinical researchers largely in the dark. Overall, the creativity, flexibility, and innovation of these modalities for solid tumor treatments are greatly encouraging, and usher in a new age of pharmaceutical development.
Collapse
|
21
|
Mamnoon B, Feng L, Froberg J, Choi Y, Sathish V, Taratula O, Taratula O, Mallik S. Targeting Estrogen Receptor-Positive Breast Microtumors with Endoxifen-Conjugated, Hypoxia-Sensitive Polymersomes. ACS OMEGA 2021; 6:27654-27667. [PMID: 34722965 PMCID: PMC8552235 DOI: 10.1021/acsomega.1c02250] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 09/20/2021] [Indexed: 06/13/2023]
Abstract
Endoxifen is the primary active metabolite of tamoxifen, a nonsteroidal-selective estrogen receptor modulator (SERM) and widely used medication to treat estrogen receptor-positive (ER+) breast cancer. In this study, endoxifen was conjugated to the surface of polymeric nanoparticles (polymersomes) for targeted delivery of doxorubicin (DOX) to estrogen receptor-positive breast cancer cells (MCF7). Rapid cell growth and insufficient blood supply result in low oxygen concentration (hypoxia) within the solid breast tumors. The polymersomes developed here are prepared from amphiphilic copolymers of polylactic acid (PLA) and poly(ethylene glycol) (PEG) containing diazobenzene as the hypoxia-responsive linker. We prepared two nanoparticle formulations: DOX-encapsulated hypoxia-responsive polymersomes (DOX-HRPs) and endoxifen-conjugated, DOX-encapsulated hypoxia-responsive polymersomes (END-DOX-HRPs). Cellular internalization studies demonstrated eight times higher cytosolic and nuclear localization after incubating breast cancer cells with END-DOX-HRPs (targeted polymersomes) in contrast to DOX-HRPs (nontargeted polymersomes). Cytotoxicity studies on monolayer cell cultures exhibited that END-DOX-HRPs were three times more toxic to ER+ MCF7 cells than DOX-HRPs and free DOX in hypoxia. The cell viability studies on three-dimensional hypoxic cultures also demonstrated twice as much toxicity when the spheroids were treated with targeted polymersomes instead of nontargeted counterparts. This is the first report of surface-decorated polymeric nanoparticles with endoxifen ligands for targeted drug delivery to ER+ breast cancer microtumors. The newly designed endoxifen-conjugated, hypoxia-responsive polymersomes might have translational potential for ER+ breast cancer treatment.
Collapse
Affiliation(s)
- Babak Mamnoon
- Department
of Pharmaceutical Sciences, North Dakota
State University, Fargo, North Dakota 58102, United States
| | - Li Feng
- Department
of Pharmaceutical Sciences, North Dakota
State University, Fargo, North Dakota 58102, United States
| | - Jamie Froberg
- Department
of Physics, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Yongki Choi
- Department
of Physics, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Venkatachalem Sathish
- Department
of Pharmaceutical Sciences, North Dakota
State University, Fargo, North Dakota 58102, United States
| | - Oleh Taratula
- Department
of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, Oregon 97201, United States
| | - Olena Taratula
- Department
of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, Oregon 97201, United States
| | - Sanku Mallik
- Department
of Pharmaceutical Sciences, North Dakota
State University, Fargo, North Dakota 58102, United States
| |
Collapse
|
22
|
Chen D, Huang H, Zang L, Gao W, Zhu H, Yu X. Development and Verification of the Hypoxia- and Immune-Associated Prognostic Signature for Pancreatic Ductal Adenocarcinoma. Front Immunol 2021; 12:728062. [PMID: 34691034 PMCID: PMC8526937 DOI: 10.3389/fimmu.2021.728062] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 09/13/2021] [Indexed: 01/02/2023] Open
Abstract
We aim to construct a hypoxia- and immune-associated risk score model to predict the prognosis of patients with pancreatic ductal adenocarcinoma (PDAC). By unsupervised consensus clustering algorithms, we generate two different hypoxia clusters. Then, we screened out 682 hypoxia-associated and 528 immune-associated PDAC differentially expressed genes (DEGs) of PDAC using Pearson correlation analysis based on the Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression project (GTEx) dataset. Seven hypoxia and immune-associated signature genes (S100A16, PPP3CA, SEMA3C, PLAU, IL18, GDF11, and NR0B1) were identified to construct a risk score model using the Univariate Cox regression and the Least Absolute Shrinkage and Selection Operator (LASSO) Cox regression, which stratified patients into high- and low-risk groups and were further validated in the GEO and ICGC cohort. Patients in the low-risk group showed superior overall survival (OS) to their high-risk counterparts (p < 0.05). Moreover, it was suggested by multivariate Cox regression that our constructed hypoxia-associated and immune-associated prognosis signature might be used as the independent factor for prognosis prediction (p < 0.001). By CIBERSORT and ESTIMATE algorithms, we discovered that patients in high-risk groups had lower immune score, stromal score, and immune checkpoint expression such as PD-L1, and different immunocyte infiltration states compared with those low-risk patients. The mutation spectrum also differs between high- and low-risk groups. To sum up, our hypoxia- and immune-associated prognostic signature can be used as an approach to stratify the risk of PDAC.
Collapse
Affiliation(s)
- Dongjie Chen
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Hui Huang
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Longjun Zang
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Wenzhe Gao
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Hongwei Zhu
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiao Yu
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
23
|
Vasantha Ramachandran R, Bhat R, Kumar Saini D, Ghosh A. Theragnostic nanomotors: Successes and upcoming challenges. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1736. [PMID: 34173342 DOI: 10.1002/wnan.1736] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/28/2021] [Accepted: 05/30/2021] [Indexed: 12/12/2022]
Abstract
The idea of "fantastic voyagers" carrying out medical tasks within the human body has existed as part of popular culture for many decades. The concept revolved around a miniaturized robot that can travel inside the human body and perform complicated functions such as surgery, navigation of otherwise inaccessible biological environments, and delivery of therapeutics. Since the last decade, significant developments have occurred in this arena that are yet to enter mainstream biomedical practises. Here, we define the challenges to make this fiction into reality. We begin by chalking the journey from pills, nanoparticles, and then to micro-nanomotors. The review describes the principles, physicochemical contexts, and advantages that micro-nanomotors provide. The article then describes micro-nanomotors' obstacles such as maneuverability, in vivo imaging, toxicity, and biodistribution. This article is categorized under: Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
| | - Ramray Bhat
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| | - Deepak Kumar Saini
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, India.,Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| | - Ambarish Ghosh
- Centre for Nano Science and Engineering, Indian Institute of Science, Bangalore, India.,Department of Physics, Indian Institute of Science, Bangalore, India
| |
Collapse
|
24
|
Bouhaoui A, Eddahmi M, Dib M, Khouili M, Aires A, Catto M, Bouissane L. Synthesis and Biological Properties of Coumarin Derivatives. A Review. ChemistrySelect 2021. [DOI: 10.1002/slct.202101346] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Abderrazzak Bouhaoui
- Organic and Analytical Chemistry Laboratory Faculty of Sciences and Technologies Sultan Moulay Slimane University BP 523 23000 Beni-Mellal Morocco
| | - Mohammed Eddahmi
- Organic and Analytical Chemistry Laboratory Faculty of Sciences and Technologies Sultan Moulay Slimane University BP 523 23000 Beni-Mellal Morocco
| | - Mustapha Dib
- Organic and Analytical Chemistry Laboratory Faculty of Sciences and Technologies Sultan Moulay Slimane University BP 523 23000 Beni-Mellal Morocco
| | - Mostafa Khouili
- Organic and Analytical Chemistry Laboratory Faculty of Sciences and Technologies Sultan Moulay Slimane University BP 523 23000 Beni-Mellal Morocco
| | - Alfredo Aires
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences CITAB University of Trás-os-Montes e Alto Douro UTAD Vila Real Portugal
| | - Marco Catto
- Department of Pharmacy-Pharmaceutical Sciences University of Bari Aldo Moro via E. Orabona 4 70125 Bari Italy
| | - Latifa Bouissane
- Organic and Analytical Chemistry Laboratory Faculty of Sciences and Technologies Sultan Moulay Slimane University BP 523 23000 Beni-Mellal Morocco
| |
Collapse
|
25
|
Dahal D, Ray P, Pan D. Unlocking the power of optical imaging in the second biological window: Structuring near-infrared II materials from organic molecules to nanoparticles. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1734. [PMID: 34159753 DOI: 10.1002/wnan.1734] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/16/2021] [Accepted: 05/24/2021] [Indexed: 12/16/2022]
Abstract
Biomedical imaging techniques play a crucial role in clinical diagnosis, surgical intervention, and prognosis. Fluorescence imaging in the second biological window (second near-infrared [NIR-II]; 1000-1700 nm) has attracted attention recently. NIR-II fluorescence imaging offers unique advantages in terms of reduced photon scattering, deep tissue penetration, high sensitivity, and many others. A host of materials, including small organic molecules, single-walled carbon nanotubes, polymeric and rare-earth-doped nanoparticles, have been explored as NIR-II emitting fluorescent probes. Efficient and viable approaches to design and develop fluorescence probes with tunable photophysical properties without compromising other key features are of paramount importance. Various chemical strategies are explored to increase the quantum yield of these imaging agents without compromising their spatiotemporal resolution, specificity, and tissue penetration capabilities. This review summarizes the strategies implemented to design and synthesize NIR-II emitting nanoparticles and small organic molecule-based fluorescent probes for applications in the biomedical field. This article is categorized under: Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Implantable Materials and Surgical Technologies > Nanoscale Tools and Techniques in Surgery.
Collapse
Affiliation(s)
- Dipendra Dahal
- Department of Pediatrics, Center for Blood Oxygen Transport and Hemostasis, University of Maryland Baltimore School of Medicine, Baltimore, Maryland, USA
| | - Priyanka Ray
- Department of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, Baltimore, Maryland, USA
| | - Dipanjan Pan
- Department of Pediatrics, Center for Blood Oxygen Transport and Hemostasis, University of Maryland Baltimore School of Medicine, Baltimore, Maryland, USA.,Department of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, Baltimore, Maryland, USA.,Department of Diagnostic Radiology and Nuclear Medicine, Center for Blood Oxygen Transport and Hemostasis, University of Maryland Baltimore School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
26
|
Dynamic cellular biomechanics in responses to chemotherapeutic drug in hypoxia probed by atomic force spectroscopy. Oncotarget 2021; 12:1165-1177. [PMID: 34136085 PMCID: PMC8202777 DOI: 10.18632/oncotarget.27974] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 05/14/2021] [Indexed: 12/11/2022] Open
Abstract
The changes in cellular structure play an important role in cancer cell development, progression, and metastasis. By exploiting single-cell, force spectroscopy methods, we probed biophysical and biomechanical kinetics (stiffness, morphology, roughness, adhesion) of brain, breast, prostate, and pancreatic cancer cells with standard chemotherapeutic drugs in normoxia and hypoxia over 12–24 hours. After exposure to the drugs, we found that brain, breast, and pancreatic cancer cells became approximately 55–75% less stiff, while prostate cancer cells became more stiff, due to either drug-induced disruption or reinforcement of cytoskeletal structure. However, the rate of the stiffness change decreased up to 2-folds in hypoxia, suggesting a correlation between cellular stiffness and drug resistance of cancer cells in hypoxic tumor microenvironment. Also, we observed significant changes in the cell body height, surface roughness, and cytoadhesion of cancer cells after exposure to drugs, which followed the trend of stiffness. Our results show that a degree of chemotherapeutic drug effects on biomechanical and biophysical properties of cancer cells is distinguishable in normoxia and hypoxia, which are correlated with alteration of cytoskeletal structure and integrity during drug-induced apoptotic process.
Collapse
|
27
|
Lou XF, Du YZ, Xu XL. Endogenous Enzyme-responsive Nanoplatforms for Anti-tumor Therapy. Curr Drug Targets 2021; 22:845-855. [PMID: 33459230 DOI: 10.2174/1389450122666210114095614] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/28/2020] [Accepted: 11/11/2020] [Indexed: 11/22/2022]
Abstract
The emergency of responsive drug delivery systems has contributed to reduced cytotoxicity, improved permeability in tissues and extended circulation time of the active drug. In particular, enzyme-responsive nanoplatforms have attracted a lot of attention due to the specificity and efficiency of an enzyme-catalyzed reaction. In this review, enzyme-based mono responsive drug delivery systems designed in the past 5 years have been summarized. These drug delivery systems were introduced by different tumor-related enzymes such as matrix metalloproteinase, esterase, hyaluronidase, caspase and cathepsin. Moreover, the enzyme-sensitive nanoplatforms activated by dual-stimuli have been also described. Although great progress had been made in the past years, the translation into clinical practice is still difficult. Thus, three obstacles (enzyme heterogeneity, reaction environment, animal model) were also discussed. In short, enzyme-activated drug delivery systems offer great potential in treating cancers.
Collapse
Affiliation(s)
- Xue-Fang Lou
- School of Medicine, Zhejiang University City College, 51 Hu-Zhou Street, Hangzhou 310015, China
| | - Yong-Zhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiao-Ling Xu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
28
|
Ray P, Moitra P, Pan D. Emerging theranostic applications of carbon dots and its variants. VIEW 2021. [DOI: 10.1002/viw.20200089] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Priyanka Ray
- Department of Chemical Biochemical, and Environmental Engineering University of Maryland Baltimore County Baltimore Maryland USA
- Department of Diagnostic Radiology and Nuclear Medicine University of Maryland Baltimore Baltimore Maryland USA
| | - Parikshit Moitra
- Department of Chemical Biochemical, and Environmental Engineering University of Maryland Baltimore County Baltimore Maryland USA
- Department of Pediatrics Center for Blood Oxygen Transport and Hemostasis University of Maryland Baltimore School of Medicine Baltimore Maryland USA
| | - Dipanjan Pan
- Department of Chemical Biochemical, and Environmental Engineering University of Maryland Baltimore County Baltimore Maryland USA
- Department of Pediatrics Center for Blood Oxygen Transport and Hemostasis University of Maryland Baltimore School of Medicine Baltimore Maryland USA
- Department of Diagnostic Radiology and Nuclear Medicine University of Maryland Baltimore Baltimore Maryland USA
| |
Collapse
|
29
|
Hu X, Xia F, Lee J, Li F, Lu X, Zhuo X, Nie G, Ling D. Tailor-Made Nanomaterials for Diagnosis and Therapy of Pancreatic Ductal Adenocarcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2002545. [PMID: 33854877 PMCID: PMC8025024 DOI: 10.1002/advs.202002545] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 10/25/2020] [Indexed: 05/05/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers worldwide due to its aggressiveness and the challenge to early diagnosis and treatment. In recent decades, nanomaterials have received increasing attention for diagnosis and therapy of PDAC. However, these designs are mainly focused on the macroscopic tumor therapeutic effect, while the crucial nano-bio interactions in the heterogeneous microenvironment of PDAC remain poorly understood. As a result, the majority of potent nanomedicines show limited performance in ameliorating PDAC in clinical translation. Therefore, exploiting the unique nature of the PDAC by detecting potential biomarkers together with a deep understanding of nano-bio interactions that occur in the tumor microenvironment is pivotal to the design of PDAC-tailored effective nanomedicine. This review will introduce tailor-made nanomaterials-enabled laboratory tests and advanced noninvasive imaging technologies for early and accurate diagnosis of PDAC. Moreover, the fabrication of a myriad of tailor-made nanomaterials for various PDAC therapeutic modalities will be reviewed. Furthermore, much preferred theranostic multifunctional nanomaterials for imaging-guided therapies of PDAC will be elaborated. Lastly, the prospects of these nanomaterials in terms of clinical translation and potential breakthroughs will be briefly discussed.
Collapse
Affiliation(s)
- Xi Hu
- Department of Clinical PharmacyZhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Researchthe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
| | - Fan Xia
- Institute of PharmaceuticsZhejiang Province Key Laboratory of Anti‐Cancer Drug ResearchHangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Jiyoung Lee
- Institute of PharmaceuticsZhejiang Province Key Laboratory of Anti‐Cancer Drug ResearchHangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Fangyuan Li
- Institute of PharmaceuticsZhejiang Province Key Laboratory of Anti‐Cancer Drug ResearchHangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
- Key Laboratory of Biomedical Engineering of the Ministry of EducationCollege of Biomedical Engineering & Instrument ScienceZhejiang UniversityHangzhou310058China
| | - Xiaoyang Lu
- Department of Clinical PharmacyZhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Researchthe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
| | - Xiaozhen Zhuo
- Department of Cardiologythe First Affiliated HospitalXi'an Jiaotong UniversityXi'an710061China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyNo.11 Zhongguancun BeiyitiaoBeijing100190China
- GBA Research Innovation Institute for NanotechnologyGuangzhou510700China
| | - Daishun Ling
- Institute of PharmaceuticsZhejiang Province Key Laboratory of Anti‐Cancer Drug ResearchHangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
- Key Laboratory of Biomedical Engineering of the Ministry of EducationCollege of Biomedical Engineering & Instrument ScienceZhejiang UniversityHangzhou310058China
| |
Collapse
|
30
|
Mamnoon B, Loganathan J, Confeld MI, De Fonseka N, Feng L, Froberg J, Choi Y, Tuvin DM, Sathish V, Mallik S. Targeted polymeric nanoparticles for drug delivery to hypoxic, triple-negative breast tumors. ACS APPLIED BIO MATERIALS 2020; 4:1450-1460. [PMID: 33954285 DOI: 10.1021/acsabm.0c01336] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
High recurrence and metastasis to vital organs are the major characteristics of triple-negative breast cancer (TNBC). Low vascular oxygen tension promotes resistance to chemo- and radiation therapy. Neuropilin-1 (NRP-1) receptor is highly expressed on TNBC cells. The tumor-penetrating iRGD peptide interacts with the NRP-1 receptor, triggers endocytosis and transcytosis, and facilitates penetration. Herein, we synthesized a hypoxia-responsive diblock PLA-diazobenzene-PEG copolymer and prepared self-assembled hypoxia-responsive polymersomes (Ps) in an aqueous buffer. The iRGD peptide was incorporated into the polymersome structure to make hypoxia-responsive iRGD-conjugated polymersomes (iPs). Doxorubicin (DOX) was encapsulated in the polymersomes to prepare both targeted and non-targeted hypoxia-responsive polymersomes (DOX-iPs and DOX-Ps, respectively). The polymeric nanoparticles released less than 30% of their encapsulated DOX within 12 hours under normoxic conditions (21% oxygen), whereas under hypoxia (2% Oxygen), doxorubicin release remarkably increased to over 95%. The targeted polymersomes significantly decreased TNBC cells' viability in monolayer and spheroid cultures under hypoxia compared to normoxia. Animal studies displayed that targeted polymersomes significantly diminished tumor growth in xenograft nude mice. Overall, the targeted polymersomes exhibited potent anti-tumor activity in monolayer, spheroid, and animal models of TNBC. With further developments, the targeted nanocarriers discussed here might have the translational potential as drug carriers for the treatment of TNBC.
Collapse
Affiliation(s)
- Babak Mamnoon
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Jagadish Loganathan
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Matthew I Confeld
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Nimesha De Fonseka
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Li Feng
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Jamie Froberg
- Department of Physics, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Yongki Choi
- Department of Physics, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Daniel M Tuvin
- Sanford Broadway Clinic, Fargo, North Dakota 58102, United States
| | - Venkatachalem Sathish
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Sanku Mallik
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58102, United States
| |
Collapse
|
31
|
Kharkar PS. Cancer Stem Cell (CSC) Inhibitors in Oncology-A Promise for a Better Therapeutic Outcome: State of the Art and Future Perspectives. J Med Chem 2020; 63:15279-15307. [PMID: 33325699 DOI: 10.1021/acs.jmedchem.0c01336] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cancer stem cells (CSCs), a subpopulation of cancer cells endowed with self-renewal, tumorigenicity, pluripotency, chemoresistance, differentiation, invasive ability, and plasticity, reside in specialized tumor niches and are responsible for tumor maintenance, metastasis, therapy resistance, and tumor relapse. The new-age "hierarchical or CSC" model of tumor heterogeneity is based on the concept of eradicating CSCs to prevent tumor relapse and therapy resistance. Small-molecular entities and biologics acting on various stemness signaling pathways, surface markers, efflux transporters, or components of complex tumor microenvironment are under intense investigation as potential anti-CSC agents. In addition, smart nanotherapeutic tools have proved their utility in achieving CSC targeting. Several CSC inhibitors in clinical development have shown promise, either as mono- or combination therapy, in refractory and difficult-to-treat cancers. Clinical investigations with CSC marker follow-up as a measure of clinical efficacy are needed to turn the "hype" into the "hope" these new-age oncology therapeutics have to offer.
Collapse
Affiliation(s)
- Prashant S Kharkar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Matunga, Mumbai 400 019, India
| |
Collapse
|
32
|
Mamnoon B, Feng L, Froberg J, Choi Y, Venkatachalem S, Mallik S. Hypoxia-Responsive, Polymeric Nanocarriers for Targeted Drug Delivery to Estrogen Receptor-Positive Breast Cancer Cell Spheroids. Mol Pharm 2020; 17:4312-4322. [PMID: 32926627 PMCID: PMC8095663 DOI: 10.1021/acs.molpharmaceut.0c00754] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Uncontrolled cell growth, division, and lack of enough blood supply causes low oxygen content or hypoxia in cancerous tumor microenvironments. 17β-Estradiol (E2), an estrogen receptor (ER) ligand, can be incorporated on the surface of nanocarriers for targeted drug delivery to breast cancer cells overexpressing ER. In the present study, we synthesized estradiol-conjugated hypoxia-responsive polymeric nanoparticles (polymersomes) encapsulating the anticancer drug doxorubicin (E2-Dox-HRPs) for targeted delivery into the hypoxic niches of estrogen-receptor-positive breast cancer microtumors. Estradiol-conjugated polymersomes released over 90% of their encapsulated Dox in a sustained manner within hypoxia (2% oxygen) after 12 h. However, they released about 30% of Dox in normal oxygen partial pressure (21% oxygen, normoxia) during this time. Fluorescence microscopic studies demonstrated higher cytosolic and nuclear internalization of E2-Dox-HRPs (targeted polymersomes) compared to those of Dox-HRPs (nontargeted polymersomes). Monolayer cell viability studies on ER-positive MCF7 cells showed higher cytotoxicity of targeted polymersomes in hypoxia compared to in normoxia. Cytotoxicity studies with hypoxic three-dimensional spheroid cultures of MCF7 cells treated with targeted polymersomes indicated significant differences compared to those of normoxic spheroids. The novel estradiol-conjugated hypoxia-responsive polymersomes described here have the potential for targeted drug delivery in estrogen-receptor-positive breast cancer therapy.
Collapse
Affiliation(s)
- Babak Mamnoon
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Li Feng
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Jamie Froberg
- Department of Physics, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Yongki Choi
- Department of Physics, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Sathish Venkatachalem
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Sanku Mallik
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58102, United States
| |
Collapse
|