Encapsulation of daunorubicin into Saccharomyces cerevisiae-derived lysosome as drug delivery vehicles for acute myeloid leukemia (AML) treatment

2020 ◽  
Vol 308 ◽  
pp. 118-123
Author(s):  
Wooil Choi ◽  
Mi Young Heo ◽  
Sang Yong Kim ◽  
Ji-Hyang Wee ◽  
Yang-Hoon Kim ◽  
...  
Author(s):  
Huiyuan Bai ◽  
Quanhao Sun ◽  
Fei Kong ◽  
Hai-Jiao Dong ◽  
Ming Ma ◽  
...  

Multifunctional drug delivery systems combining two or more therapies have broad prospects for high efficacy tumor treatment. Herein, we designed a novel hollow mesoporous Prussian blue nanoparticles (HMPBs)-based platform for...


Genetics ◽  
1999 ◽  
Vol 153 (1) ◽  
pp. 13-23 ◽  
Author(s):  
Eugenia Y Xu ◽  
Susan Kim ◽  
Kirstin Replogle ◽  
Jasper Rine ◽  
David H Rivier

Abstract In Saccharomyces cerevisiae, chromatin-mediated silencing inactivates transcription of the genes at the HML and HMR cryptic mating-type loci and genes near telomeres. Mutations in the Rap1p and Abf1p binding sites of the HMR-E silencer (HMRa-e**) result in a loss of silencing at HMR. We characterized a collection of 15 mutations that restore the α-mating phenotype to MATα HMRa-e** strains. These mutations defined three complementation groups, two new groups and one group that corresponded to the previously identified SAS2 gene. We cloned the genes that complemented members of the new groups and identified two previously uncharacterized genes, which we named SAS4 and SAS5. Neither SAS4 nor SAS5 was required for viability. Null alleles of SAS4 and SAS5 restored SIR4-dependent silencing at HMR, establishing that each is a regulator of silencing. Null alleles of SAS4 and SAS5 bypassed the role of the Abf1p binding site of the HMR-E silencer but not the role of the ACS or Rap1p binding site. Previous analysis indicated that SAS2 is homologous to a human gene that is a site of recurring translocations involved in acute myeloid leukemia. Similarly, SAS5 is a member of a gene family that included two human genes that are the sites of recurring translocations involved in acute myeloid leukemia.


2020 ◽  
Vol 6 (50) ◽  
pp. eabc3013
Author(s):  
Tianyuan Ci ◽  
Hongjun Li ◽  
Guojun Chen ◽  
Zejun Wang ◽  
Jinqiang Wang ◽  
...  

Live cells have been vastly engineered into drug delivery vehicles to leverage their targeting capability and cargo release behavior. Here, we describe a simple method to obtain therapeutics-containing “dead cells” by shocking live cancer cells in liquid nitrogen to eliminate pathogenicity while preserving their major structure and chemotaxis toward the lesion site. In an acute myeloid leukemia (AML) mouse model, we demonstrated that the liquid nitrogen–treated AML cells (LNT cells) can augment targeted delivery of doxorubicin (DOX) toward the bone marrow. Moreover, LNT cells serve as a cancer vaccine and promote antitumor immune responses that prolong the survival of tumor-bearing mice. Preimmunization with LNT cells along with an adjuvant also protected healthy mice from AML cell challenge.


2017 ◽  
Vol 13 (5) ◽  
pp. 500-512 ◽  
Author(s):  
Kheireddine El-Boubbou ◽  
Daniel Azar ◽  
Amira Bekdash ◽  
Ralph J. Abi-Habib

2019 ◽  
Vol 27 (1-2) ◽  
pp. 45-55 ◽  
Author(s):  
Yiqian Wang ◽  
Ying Xie ◽  
Jacob Williams ◽  
Yu Hang ◽  
Lisa Richter ◽  
...  

2008 ◽  
Vol 5 (6) ◽  
pp. 653-663 ◽  
Author(s):  
Johannes Kohlschütter ◽  
Stefan Michelfelder ◽  
Martin Trepel

Molecules ◽  
2019 ◽  
Vol 24 (11) ◽  
pp. 2103 ◽  
Author(s):  
Noureldien H. E. Darwish ◽  
Thangirala Sudha ◽  
Kavitha Godugu ◽  
Dhruba J. Bharali ◽  
Osama Elbaz ◽  
...  

The targeted nano-encapsulation of anticancer drugs can improve drug delivery and the selective targeting of cancer cells. Nuclear factor kappa B (NF-kB) is a regulator for different biological responses, including cell proliferation and differentiation. In acute myeloid leukemia (AML), constitutive NF-κB has been detected in more than 50% of cases, enabling leukemic cells to resist apoptosis and stimulate uncontrolled proliferation. We evaluated NF-kB expression in bone marrow samples from 103 patients with AML using quantitative real time polymerase chain reaction (RT-PCR) and found that expression was increased in 80.5% (83 out 103) of these patients with AML in comparison to the control group. Furthermore, overexpressed transmembrane glycoprotein (CD44) on leukemic cells in comparison to normal cells is known to play an important role in leukemic cell engraftment and survival. We designed poly lactide co-glycolide (PLGA) nanoparticles conjugated with antiCD44 and encapsulating parthenolide (PTL), a nuclear factor kappa B (NF-kB) inhibitor, in order to improve the selectivity and targeting of leukemic cells and to spare normal cells. In vitro, in leukemic cell lines Kasumi-1, KG-1a, and THP-1, proliferation was decreased by 40% (** p < 0.01) with 5 µM PLGA-antiCD44-PTL nanoparticles in comparison to the same concentration of free PTL (~10%). The higher uptake of the nanoparticles by leukemic cells was confirmed with confocal microscopy. In conclusion, PLGA-antiCD44-PTL nanoparticles improved the bioavailability and selective targeting of leukemic cells, thus holding promise as a drug delivery system to improve the cure rate of AML.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3595-3595 ◽  
Author(s):  
Rose McGlauflin ◽  
Pan Li ◽  
Prudovsky Igor ◽  
Calvin Vary ◽  
Pradeep Sathyanarayana

Abstract Background: Exosomes are microvesicles that play important roles in intercellular communications in both normal and tumor cells via their cargoes, which include microRNA (miRNA) and proteins. miRNAs play critical regulatory roles in hematopoiesis, and their abnormal expression is correlated with several hematological malignancies, including acute myeloid leukemia (AML). Exosome-derived miRNAs and proteins are increasingly recognized for their prognostic biomarker potential. Exosomes are being evaluated for their potential as novel drug-delivery vehicles due to their endogenous nature and ability to carry small molecule drugs. Aims: We aim to characterize HSC-derived exosomes, investigate the biomarker potential of exosomes derived from AML patient samples (by examining their proteomic and miRNA profiles) and utilize exosomes as innovative drug delivery vehicles with the ability to eliminate leukemic stem cells in a targeted manner. Methods: Secreted exosomes from murine bone marrow HSCs were isolated from conditioned medium and visualized using confocal microscopy. We isolated exosomes and performed miRNA profiling, using qPCR, and LC-MS/MS proteomic analysis to characterize the constituents. We also isolated exosomes from the CD34+ cells of three AML patient samples and profiled 372 of the most abundantly expressed miRs in these cells compared to normal CD34+ cells. We analyzed the proteome of these exosomes as well. In order to assess the utility of exosomes as drug carriers, exosomes from bone marrow-derived OP9 stromal cells were transfected with Daunorubicin (1ug/ul). Normal CD34+ cells and patient-derived AML samples (from n=2 patients) were treated with varying doses of the drug-loaded exosomes. Drug-loaded exosomes uptake was tracked with a Texas Red siRNA. After 24 hours, cells were screened for apoptosis. To test the feasibility of targeted exosomes, OP9 cells were exposed to AML patient samples for 48 hours. The patient cells were then removed and, 24 hours later, "trained" stromal cell-derived exosomes were isolated from the media and transfected with Daunorubicin. These patient-specific, exosomes were plated with both the corresponding patient's CD34+ cells as well as normal CD34+ primary cells. After 24 hours, apoptosis was measured. Results: miRNA profiling of murine bone marrow showed miR-21a, miR-92a and miR-25 were most abundant in exosomes. Proteomic LC-MS/MS analysis revealed presence of exosome-associated novel proteins such as Syntenin-1. Syntenin-1, which is known to bind IL-5R and promote myelopoiesis, was present in significantly higher levels in HSCs compared to myeloid progenitors, implying a functional role for exosome derived Syntenin-1. miRNA profiling in AML samples revealed distinct signature profiles. Importantly, exosome derived miRs such as -1290, -375, -205 and -21-that are known prognostic markers in cancers such as prostate, ovarian and hepatocellular carcinoma-were significantly upregulated in all the three exosome-derived AML samples. The drug-loaded exosomes were successful in inducing significant apoptosis in two patient samples tested. These drug-loaded exosomes also induced cell death in CD34+ normal cells when compared to control exosomes. However, the patient-trained exosomes specifically eliminated 92% of CD34+ AML patient cells, while causing significantly less cell death (44%) of normal CD34+ primary cells exposed to drug-loaded, patient-trained exosomes. Summary/Conclusion: Taken together, our data predict important functional roles for exosome-derived Syntenin-1 in regulating lineage specific hematopoietic differentiations. Furthermore, for the first time, we have identified highly upregulated select exosome-derived miRs from AML patient samples whose prognostic value has been recently reported for other cancers, making these miRs promising candidates for AML biomarkers as well. Finally, stromal cell-derived, drug-loaded exosomes are not only able to induce apoptosis in AML patient samples, but they can effectively be trained by leukemic cells to favor uptake resulting in targeted elimination of leukemic over normal CD34+ cells. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document