1
|
Sun J, Li HL, Zhou WJ, Ma ZX, Huang XP, Li C. Current status and recent progress of nanomaterials in transcatheter arterial chemoembolization therapy for hepatocellular carcinoma. World J Clin Oncol 2025; 16:104435. [PMID: 40290691 PMCID: PMC12019268 DOI: 10.5306/wjco.v16.i4.104435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/06/2025] [Accepted: 03/05/2025] [Indexed: 03/26/2025] Open
Abstract
Hepatocellular carcinoma (HCC) remains one of the most common cancers worldwide. Transcatheter arterial chemoembolization has become a common treatment modality for some patients with unresectable advanced HCC. Since the introduction of nanomaterials in 1974, their use in various fields has evolved rapidly. In medical applications, nanomaterials can serve as carriers for the delivery of chemotherapeutic drugs to tumour tissues. Additionally, nanomaterials have potential for in vivo tumour imaging. This article covers the properties and uses of several kinds of nanomaterials, focusing on their use in transcatheter arterial chemoembolization for HCC treatment. This paper also discusses the limitations currently associated with the use of nanomaterials.
Collapse
Affiliation(s)
- Jia Sun
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, Guangdong Province, China
| | - Hai-Liang Li
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, Guangdong Province, China
| | - Wen-Jun Zhou
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, Guangdong Province, China
| | - Zeng-Xin Ma
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, Guangdong Province, China
| | - Xiao-Pei Huang
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, Guangdong Province, China
| | - Cheng Li
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, Guangdong Province, China
| |
Collapse
|
2
|
Mohammadian J, Mahmoudi S, Pourmohammad P, Pirouzpanah M, Salehnia F, Maroufi NF, Samadi N, Sabzichi M. Formulation of Stattic as STAT3 inhibitor in nanostructured lipid carriers (NLCs) enhances efficacy of doxorubicin in melanoma cancer cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2020; 393:2315-2323. [PMID: 32653978 DOI: 10.1007/s00210-020-01942-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/03/2020] [Indexed: 01/29/2023]
Abstract
Nowadays, nanoparticle-based combination therapy has been emerging as huge innovation in cancer treatment. Here, we studied the effect of Stattic (STAT3 inhibitor) loaded in nanostructured lipid carriers (NLCs) on enhancing the efficacy, cytotoxicity, and induction of apoptosis of doxorubicin in B16F10 mouse melanoma cancer cell. The evaluation of Stattic-loaded NLCs has been done in terms of zeta potential, particle size, scanning electron microscope (SEM), and cellular uptake. MTT assay was applied to evaluate the cell proliferation. Apoptotic cell death and identification of early and late apoptosis were assessed by DAPI staining and Annexin V/PI staining, respectively. Real-time RT-PCR was applied to measure the effects of doxorubicin and/or Stattic on key apoptotic genes such as Bad, Survivin, HIF1, and STAT3. The Stattic formulated into NLCs shown mean particle size of 56 ± 7 nm which was confirmed by SEM. The IC50 values for Stattic and doxorubicin were 2.95 ± 0.52 μM and 1.21 ± 0.36 μM, respectively. Stattic-loaded NLCs diminished percent of cell proliferation from 68 ± 6.8 to 54 ± 3.7% (p < 0.05). Combinational treatment of the cells with Stattic-loaded nanoparticles and doxorubicin give rise to a significant increase in the percentage of apoptosis (p < 0.05). The study of gene expression profile has shown a remarkable decrease in anti-apoptotic gene, Survivin, along with smooth decline in HIF1 as angiogenesis intermediator and increase in Bad mRNA levels. Our results recommend that NLCs as novel technology have potent strategy to augment efficacy of current chemotherapeutic agent in melanoma cancer cells.
Collapse
Affiliation(s)
- Jamal Mohammadian
- Drug Applied Research Center, and Department of Medical Biotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shiva Mahmoudi
- Drug Applied Research Center, and Department of Medical Biotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Clinical Biochemistry, School of Medicine, Ardabil University of Medical Science, Ardabil, Iran
| | - Pirouz Pourmohammad
- Department of Clinical Biochemistry, School of Medicine, Ardabil University of Medical Science, Ardabil, Iran
| | - Mohammad Pirouzpanah
- Department of Genetic and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, 34755, Istanbul, Turkey
| | - Fatemeh Salehnia
- Drug Applied Research Center, and Department of Medical Biotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Fathi Maroufi
- Drug Applied Research Center, and Department of Medical Biotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasser Samadi
- Drug Applied Research Center, and Department of Medical Biotechnology, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mehdi Sabzichi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
3
|
Xu X, Liu A, Bai Y, Li Y, Zhang C, Cui S, Piao Y, Zhang S. Co-delivery of resveratrol and p53 gene via peptide cationic liposomal nanocarrier for the synergistic treatment of cervical cancer and breast cancer cells. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2018.05.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
4
|
Pugliese E, Coentro JQ, Zeugolis DI. Advancements and Challenges in Multidomain Multicargo Delivery Vehicles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1704324. [PMID: 29446161 DOI: 10.1002/adma.201704324] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/05/2017] [Indexed: 06/08/2023]
Abstract
Reparative and regenerative processes are well-orchestrated temporal and spatial events that are governed by multiple cells, molecules, signaling pathways, and interactions thereof. Yet again, currently available implantable devices fail largely to recapitulate nature's complexity and sophistication in this regard. Herein, success stories and challenges in the field of layer-by-layer, composite, self-assembly, and core-shell technologies are discussed for the development of multidomain/multicargo delivery vehicles.
Collapse
Affiliation(s)
- Eugenia Pugliese
- Regenerative, Modular and Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Ireland
- Science Foundation Ireland (SFI), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Ireland
| | - João Q Coentro
- Regenerative, Modular and Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Ireland
- Science Foundation Ireland (SFI), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Ireland
| | - Dimitrios I Zeugolis
- Regenerative, Modular and Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Ireland
- Science Foundation Ireland (SFI), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Ireland
| |
Collapse
|
5
|
Di Paolo D, Pastorino F, Brignole C, Marimpietri D, Loi M, Ponzoni M, Pagnan G. Drug Delivery Systems: Application of Liposomal Anti-Tumor Agents to Neuroectodermal Cancer Treatment. TUMORI JOURNAL 2018; 94:246-53. [DOI: 10.1177/030089160809400217] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Disseminated neuroectoderma-derived tumors, mainly neuroblastoma in childhood and melanoma in the adulthood, are refractory to most current therapeutic regimens and hence the prognosis remains very poor. Preclinical research studies have indicated several agents that show promising therapeutic potential for these neoplasms. However, there appears to be a limitation to their in vivo applicability, mainly due to unfavorable pharmacokinetic properties that lead to insufficient drug delivery to the tumor or metastatic sites or to high systemic or organ-specific toxicity. In this scenario, the focus is on targeted cancer therapy. Encapsulating anticancer drugs in liposomes enables targeted drug delivery to tumor tissue and prevents damage to the normal surrounding tissue. Indeed, sterically stabilized liposomes have been shown to enhance the selective localization of entrapped drugs to solid tumors, with improvements in therapeutic indices. The identification of tumor-associated antigens and/or genes and the relative ease of manipulating the physicochemical features of liposome hold promise for the development of novel therapeutic strategies that selectively target tumor cells. Combined targeting is still investigated, especially the availability to simultaneously target and kill both the cancer cells and the tumor vasculature. Animal models make it possible to link molecular genetics and biochemistry information to the physiological basis of disease and are important predictive tools that offer a frontline testing system for studying the involvement of specific genes and the efficacy of novel therapeutics approaches. Relevant experimental models of human neuroblastoma and melanoma, which better reflect the tumor behavior in patients, are required to evaluate the effectiveness of the various targeted liposomal formulations and their possible systemic and organ-specific toxicity. The most multifunctional targeted liposomes are herein described, with primary attention on testing their efficacy in clinically relevant animal models for the treatment of neuroblastoma and melanoma.
Collapse
Affiliation(s)
- Daniela Di Paolo
- Experimental Therapies Unit, Laboratory of Oncology, G. Gaslini Children's Hospital, Genoa, Italy
| | - Fabio Pastorino
- Experimental Therapies Unit, Laboratory of Oncology, G. Gaslini Children's Hospital, Genoa, Italy
| | - Chiara Brignole
- Experimental Therapies Unit, Laboratory of Oncology, G. Gaslini Children's Hospital, Genoa, Italy
| | - Danilo Marimpietri
- Experimental Therapies Unit, Laboratory of Oncology, G. Gaslini Children's Hospital, Genoa, Italy
| | - Monica Loi
- Experimental Therapies Unit, Laboratory of Oncology, G. Gaslini Children's Hospital, Genoa, Italy
| | - Mirco Ponzoni
- Experimental Therapies Unit, Laboratory of Oncology, G. Gaslini Children's Hospital, Genoa, Italy
| | - Gabriella Pagnan
- Experimental Therapies Unit, Laboratory of Oncology, G. Gaslini Children's Hospital, Genoa, Italy
| |
Collapse
|
6
|
Kheirolomoom A, Kim CW, Seo JW, Kumar S, Son DJ, Gagnon MKJ, Ingham ES, Ferrara KW, Jo H. Multifunctional Nanoparticles Facilitate Molecular Targeting and miRNA Delivery to Inhibit Atherosclerosis in ApoE(-/-) Mice. ACS NANO 2015; 9:8885-97. [PMID: 26308181 PMCID: PMC4581466 DOI: 10.1021/acsnano.5b02611] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 08/26/2015] [Indexed: 05/18/2023]
Abstract
The current study presents an effective and selective multifunctional nanoparticle used to deliver antiatherogenic therapeutics to inflamed pro-atherogenic regions without off-target changes in gene expression or particle-induced toxicities. MicroRNAs (miRNAs) regulate gene expression, playing a critical role in biology and disease including atherosclerosis. While anti-miRNA are emerging as therapeutics, numerous challenges remain due to their potential off-target effects, and therefore the development of carriers for selective delivery to diseased sites is important. Yet, co-optimization of multifunctional nanoparticles with high loading efficiency, a hidden cationic domain to facilitate lysosomal escape and a dense, stable incorporation of targeting moieties is challenging. Here, we create coated, cationic lipoparticles (CCLs), containing anti-miR-712 (∼1400 molecules, >95% loading efficiency) within the core and with a neutral coating, decorated with 5 mol % of peptide (VHPK) to target vascular cell adhesion molecule 1 (VCAM1). Optical imaging validated disease-specific accumulation as anti-miR-712 was efficiently delivered to inflamed mouse aortic endothelial cells in vitro and in vivo. As with the naked anti-miR-712, the delivery of VHPK-CCL-anti-miR-712 effectively downregulated the d-flow induced expression of miR-712 and also rescued the expression of its target genes tissue inhibitor of metalloproteinase 3 (TIMP3) and reversion-inducing-cysteine-rich protein with kazal motifs (RECK) in the endothelium, resulting in inhibition of metalloproteinase activity. Moreover, an 80% lower dose of VHPK-CCL-anti-miR-712 (1 mg/kg dose given twice a week), as compared with naked anti-miR-712, prevented atheroma formation in a mouse model of atherosclerosis. While delivery of naked anti-miR-712 alters expression in multiple organs, miR-712 expression in nontargeted organs was unchanged following VHPK-CCL-anti-miR-712 delivery.
Collapse
Affiliation(s)
- Azadeh Kheirolomoom
- Department of Biomedical Engineering, University of California, Davis, California 95616, United States
| | - Chan Woo Kim
- Wallace H. Coulter Department of Biomedical Engineering and Division of Cardiology, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| | - Jai Woong Seo
- Department of Biomedical Engineering, University of California, Davis, California 95616, United States
| | - Sandeep Kumar
- Wallace H. Coulter Department of Biomedical Engineering and Division of Cardiology, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| | - Dong Ju Son
- Wallace H. Coulter Department of Biomedical Engineering and Division of Cardiology, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| | - M. Karen J. Gagnon
- Department of Biomedical Engineering, University of California, Davis, California 95616, United States
| | - Elizabeth S. Ingham
- Department of Biomedical Engineering, University of California, Davis, California 95616, United States
| | - Katherine W. Ferrara
- Department of Biomedical Engineering, University of California, Davis, California 95616, United States
- Address correspondence to ,
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering and Division of Cardiology, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
- Address correspondence to ,
| |
Collapse
|
7
|
Fico A, Alfano D, Valentino A, Vasta V, Cavalcanti E, Travali S, Patriarca EJ, Caputo E. c-Myc modulation: a key role in melanoma drug response. Cancer Biol Ther 2015; 16:1375-86. [PMID: 25835050 DOI: 10.1080/15384047.2015.1030546] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Understanding molecular mechanisms involved in melanoma resistance to drugs is a big challenge. Experimental evidences suggested a correlation between mutational status in B-RAF and melanoma cell susceptibility to drugs, such as paclitaxel, doxorubicin and temozolomide, which generate an accumulation of hydrogen peroxide (H2O2) in the cells. We investigated the survival phenotype and the protein level of c-myc, a B-RAF target molecule, in melanoma cells, carrying a different mutational status in B-RAF, upon paclitaxel, doxorubicin and H2O2 treatment. For the first time, we reported c-myc modulation is critical for melanoma drug response. It appeared drug-specific and post-transcriptionally driven through PP2A; in correlation, cell pre-treatment with okadaic acid (OA), a specific PP2A inhibitor, as well as PP2A silencing of melanoma cells, was able to increase melanoma cell drug-sensitivity and c-myc protein level. This is relevant for designing efficacious therapeutic strategies in melanoma.
Collapse
Affiliation(s)
- Annalisa Fico
- a Institute of Genetics and Biophysics; A Buzzati-Traverso ; Naples , Italy.,d These authors equally contributed to this work
| | - Daniela Alfano
- a Institute of Genetics and Biophysics; A Buzzati-Traverso ; Naples , Italy.,d These authors equally contributed to this work
| | - Anna Valentino
- a Institute of Genetics and Biophysics; A Buzzati-Traverso ; Naples , Italy
| | - Valeria Vasta
- b Università degli Studi di Catania; Dipartimento di Scienze Bio-Mediche ; Catania , Italy
| | | | - Salvatore Travali
- b Università degli Studi di Catania; Dipartimento di Scienze Bio-Mediche ; Catania , Italy
| | | | - Emilia Caputo
- a Institute of Genetics and Biophysics; A Buzzati-Traverso ; Naples , Italy.,b Università degli Studi di Catania; Dipartimento di Scienze Bio-Mediche ; Catania , Italy
| |
Collapse
|
8
|
Sasi SP, Song J, Park D, Enderling H, McDonald JT, Gee H, Garrity B, Shtifman A, Yan X, Walsh K, Natarajan M, Kishore R, Goukassian DA. TNF-TNFR2/p75 signaling inhibits early and increases delayed nontargeted effects in bone marrow-derived endothelial progenitor cells. J Biol Chem 2014; 289:14178-93. [PMID: 24711449 DOI: 10.1074/jbc.m114.567743] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
TNF-α, a pro-inflammatory cytokine, is highly expressed after being irradiated (IR) and is implicated in mediating radiobiological bystander responses (RBRs). Little is known about specific TNF receptors in regulating TNF-induced RBR in bone marrow-derived endothelial progenitor cells (BM-EPCs). Full body γ-IR WT BM-EPCs showed a biphasic response: slow decay of p-H2AX foci during the initial 24 h and increase between 24 h and 7 days post-IR, indicating a significant RBR in BM-EPCs in vivo. Individual TNF receptor (TNFR) signaling in RBR was evaluated in BM-EPCs from WT, TNFR1/p55KO, and TNFR2/p75KO mice, in vitro. Compared with WT, early RBR (1-5 h) were inhibited in p55KO and p75KO EPCs, whereas delayed RBR (3-5 days) were amplified in p55KO EPCs, suggesting a possible role for TNFR2/p75 signaling in delayed RBR. Neutralizing TNF in γ-IR conditioned media (CM) of WT and p55KO BM-EPCs largely abolished RBR in both cell types. ELISA protein profiling of WT and p55KO EPC γ-IR-CM over 5 days showed significant increases in several pro-inflammatory cytokines, including TNF-α, IL-1α (Interleukin-1 alpha), RANTES (regulated on activation, normal T cell expressed and secreted), and MCP-1. In vitro treatments with murine recombinant (rm) TNF-α and rmIL-1α, but not rmMCP-1 or rmRANTES, increased the formation of p-H2AX foci in nonirradiated p55KO EPCs. We conclude that TNF-TNFR2 signaling may induce RBR in naïve BM-EPCs and that blocking TNF-TNFR2 signaling may prevent delayed RBR in BM-EPCs, conceivably, in bone marrow milieu in general.
Collapse
Affiliation(s)
- Sharath P Sasi
- From the Cardiovascular Research Center, Steward Research and Specialty Projects Corporation, Brighton, Massachusetts 02135
| | - Jin Song
- From the Cardiovascular Research Center, Steward Research and Specialty Projects Corporation, Brighton, Massachusetts 02135
| | - Daniel Park
- From the Cardiovascular Research Center, Steward Research and Specialty Projects Corporation, Brighton, Massachusetts 02135
| | - Heiko Enderling
- the Center of Cancer Systems Biology, GeneSys Research Institute, Boston, Massachusetts 02135, Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - J Tyson McDonald
- the Center of Cancer Systems Biology, GeneSys Research Institute, Boston, Massachusetts 02135, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Hannah Gee
- From the Cardiovascular Research Center, Steward Research and Specialty Projects Corporation, Brighton, Massachusetts 02135
| | - Brittany Garrity
- From the Cardiovascular Research Center, Steward Research and Specialty Projects Corporation, Brighton, Massachusetts 02135
| | - Alexander Shtifman
- From the Cardiovascular Research Center, Steward Research and Specialty Projects Corporation, Brighton, Massachusetts 02135, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Xinhua Yan
- From the Cardiovascular Research Center, Steward Research and Specialty Projects Corporation, Brighton, Massachusetts 02135, the Center of Cancer Systems Biology, GeneSys Research Institute, Boston, Massachusetts 02135, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Kenneth Walsh
- the Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Mohan Natarajan
- the University of Texas Health Science Center, San Antonio, Texas 78229, and
| | - Raj Kishore
- the Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, Illinois 60611
| | - David A Goukassian
- From the Cardiovascular Research Center, Steward Research and Specialty Projects Corporation, Brighton, Massachusetts 02135, Tufts University School of Medicine, Boston, Massachusetts 02111, the Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts 02118,
| |
Collapse
|
9
|
Yuan Y, Cai H, Yang XJ, Li W, He J, Guo TK, Chen YR. Liposome-mediated induction of apoptosis of human hepatoma cells by c-myc antisense phosphorothioate oligodeoxynucleotide and 5-fluorouracil. Asian Pac J Cancer Prev 2014; 15:5529-5533. [PMID: 25081659 DOI: 10.7314/apjcp.2014.15.14.5529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The aim of this study was to investigate the effect of a c-myc antisense oligodeoxynucleotide and 5-fluorouracil on the expression of c-myc, invasion and proliferation of HEPG-2 liver cancer cells. MATERIALS AND METHODS HEPG-2 cells were treated with lipiosome-mediated c-myc ADSON and 5-fluorouracil. The proliferation inhibition rate and invasion were measured by MTT and invasion assay, respectively. Cell apoptosis was detected by flow cytometry and expression of c-myc by RT-PCR and immunohistochemistry. RESULTS The proliferation inhibition rate was significantly higher in the antisense oligodeoxynucleotide added-5-fluorouracil group than single antisense oligodeoxynucleotide or 5-fluorouracil group (p<0.05). G0/G1 cells in the antisense oligodeoxynucleotide group and S cells in the 5-fluorouracil groups were significantly increased than that in the control group, respectively (P<0.01). The amplification strips of PCR products in 5-FU, ASODN and combination groups were significantly weaker than that in the control group (P<0.01). The percentage of c-myc-protein- positive cells were significantly lower in antisense oligodeoxynucleotide, 5-fluorouracil and combination groups than that in the control group (P<0.01). CONCLUSIONS A liposome-mediated c-myc antisense oligodeoxynucleotide and 5-fluorouracil can inhibit the proliferation and invasion of liver cancer cells by reducing the expression of c-myc. A c-myc antisense oligodeoxynucleotide can increase the sensitivity of liver cancer cells to 5-fluorouracil and decrease the dosage of the agent necessary for efficacy, providing an experimental basis for the clinical therapy of liver cancer.
Collapse
Affiliation(s)
- Yuan Yuan
- The First Hospital of Lanzhou University, Lanzhou, China E-mail :
| | | | | | | | | | | | | |
Collapse
|
10
|
Allen TM, Cullis PR. Liposomal drug delivery systems: from concept to clinical applications. Adv Drug Deliv Rev 2013; 65:36-48. [PMID: 23036225 DOI: 10.1016/j.addr.2012.09.037] [Citation(s) in RCA: 3106] [Impact Index Per Article: 258.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 09/10/2012] [Accepted: 09/20/2012] [Indexed: 02/06/2023]
Abstract
The first closed bilayer phospholipid systems, called liposomes, were described in 1965 and soon were proposed as drug delivery systems. The pioneering work of countless liposome researchers over almost 5 decades led to the development of important technical advances such as remote drug loading, extrusion for homogeneous size, long-circulating (PEGylated) liposomes, triggered release liposomes, liposomes containing nucleic acid polymers, ligand-targeted liposomes and liposomes containing combinations of drugs. These advances have led to numerous clinical trials in such diverse areas as the delivery of anti-cancer, anti-fungal and antibiotic drugs, the delivery of gene medicines, and the delivery of anesthetics and anti-inflammatory drugs. A number of liposomes (lipidic nanoparticles) are on the market, and many more are in the pipeline. Lipidic nanoparticles are the first nanomedicine delivery system to make the transition from concept to clinical application, and they are now an established technology platform with considerable clinical acceptance. We can look forward to many more clinical products in the future.
Collapse
|
11
|
Weecharangsan W, Lee RJ. Growth inhibition and chemosensitization of human carcinoma cells by human serum albumin-coated liposomal antisense oligodeoxyribonucleotide against bcl-2. Drug Deliv 2012; 19:292-7. [PMID: 22931245 DOI: 10.3109/10717544.2012.714810] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Previous study has shown human serum albumin (HSA) coated liposomes can deliver bcl-2 antisense oligodeoxyribonucleotide (ODN) into KB carcinoma cells, and decrease bcl-2 mRNA and protein expression level. In the current study, cell growth inhibition and chemosensitization of KB cells were evaluated. Liposomes composed of dimethyldioctadecyl ammonium bromide/egg phosphatidylcholine/α-tocopheryl polyethylene glycol 1000 succinate (58:40:2 molar ratio) complexed with bcl-2 antisense ODN and coated with HSA were examined for cell growth inhibition and sensitization to a commonly used chemotherapeutic drug, doxorubicin. HSA-coated liposome-ODN complexes effectively inhibited cell growth in the range of ODN concentration of 0.45-7.2 µM. Upon posttreatment with doxorubicin, the cytotoxicity was further significantly increased compared to the ODN complexes alone. The cytotoxicity was dependent on antisense ODN concentration, incubation time and doxorubicin concentration, and relatively independent on HSA concentration. This study suggests that HSA-coated liposomes are effective delivery vehicles for antisense ODN with potential therapeutic application and can be effectively combined with doxorubicin.
Collapse
Affiliation(s)
- Wanlop Weecharangsan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Srinakharinwirot University, Nakhonnayok, Thailand.
| | | |
Collapse
|
12
|
Lehmann FM, Feicht S, Helm F, Maurberger A, Ladinig C, Zimber-Strobl U, Kühn R, Mautner J, Gerbitz A, Bornkamm GW. Humanized c-Myc mouse. PLoS One 2012; 7:e42021. [PMID: 22860051 PMCID: PMC3409231 DOI: 10.1371/journal.pone.0042021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 07/02/2012] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND A given tumor is usually dependent on the oncogene that is activated in the respective tumor entity. This phenomenon called oncogene addiction provides the rationale for attempts to target oncogene products in a therapeutic manner, be it by small molecules, by small interfering RNAs (siRNA) or by antigen-specific T cells. As the proto-oncogene product is required also for the function of normal cells, this raises the question whether there is a therapeutic window between the adverse effects of specific inhibitors or T cells to normal tissue that may limit their application, and their beneficial tumor-specific therapeutic action. To address this crucial question, suitable mouse strains need to be developed, that enable expression of the human proto-oncogene not only in tumor but also in normal cells. The aim of this work is to provide such a mouse strain for the human proto-oncogene product c-MYC. PRINCIPAL FINDINGS We generated C57BL/6-derived embryonic stem cells that are transgenic for a humanized c-Myc gene and established a mouse strain (hc-Myc) that expresses human c-MYC instead of the murine ortholog. These transgenic animals harbor the humanized c-Myc gene integrated into the endogenous murine c-Myc locus. Despite the lack of the endogenous murine c-Myc gene, homozygous mice show a normal phenotype indicating that human c-MYC can replace its murine ortholog. CONCLUSIONS The newly established hc-Myc mouse strain provides a model system to study in detail the adverse effects of therapies that target the human c-MYC protein. To mimic the clinical situation, hc-Myc mice may be cross-bred to mice that develop tumors due to overexpression of human c-MYC. With these double transgenic mice it will be possible to study simultaneously the therapeutic efficiency and adverse side effects of MYC-specific therapies in the same mouse.
Collapse
Affiliation(s)
- Frank M. Lehmann
- Institute of Clinical Molecular Biology and Tumor Genetics, Helmholtz Center Munich, Munich, Germany
| | - Samantha Feicht
- Department of Gene Vectors, Helmholtz Center Munich, Munich, Germany
| | - Florian Helm
- Department of Immunology, Charité Berlin, Berlin, Germany
| | - Anna Maurberger
- Department of Hematology/Oncology, University of Erlangen, Erlangen, Germany
| | - Camilla Ladinig
- Institute of Clinical Molecular Biology and Tumor Genetics, Helmholtz Center Munich, Munich, Germany
| | | | - Ralf Kühn
- Institute of Developmental Genetics, Helmholtz Center Munich, Neuherberg, Germany
| | - Josef Mautner
- Department of Pediatrics, Technical University (TU) Munich and Clinical Cooperation Group Pediatric Tumor Immunology, TU Munich and Helmholtz Center Munich, Munich, Germany
| | - Armin Gerbitz
- Department of Hematology/Oncology, University of Erlangen, Erlangen, Germany
| | - Georg W. Bornkamm
- Institute of Clinical Molecular Biology and Tumor Genetics, Helmholtz Center Munich, Munich, Germany
- * E-mail:
| |
Collapse
|
13
|
Zapata-Benavides P, Manilla-Muñoz E, Zamora-Avila DE, Saavedra-Alonso S, Franco-Molina MA, Trejo-Avila LM, Davalos-Aranda G, Rodríguez-Padilla C. WT1 silencing by RNAi synergizes with chemotherapeutic agents and induces chemosensitization to doxorubicin and cisplatin in B16F10 murine melanoma cells. Oncol Lett 2012; 3:751-755. [PMID: 22740987 DOI: 10.3892/ol.2012.578] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Accepted: 11/17/2011] [Indexed: 12/20/2022] Open
Abstract
The Wilm's tumor gene (WT1), encoding a transcription factor that modulates the expression of certain genes that are involved in proliferation and apoptosis, is overexpressed in numerous solid tumors. WT1 is important for cell proliferation and in the diagnosis of melanoma. The objectives of this study were to investigate whether WT1 silencing is capable of synergizing with chemotherapeutic agents and whether this silencing is capable of sensitizing cancer cells to doxorubicin and cisplatin in the B16F10 murine melanoma cell line. In the present study, B16F10 cells were simultaneously treated with median lethal doses (LD50s) of WT1-1 or WT1-2 small hairpin RNAs (shRNAs) and chemotherapeutic agents. A total of 24 h post-transfection, a [3-(4,5-dimethylthiazol-2yl)-2,5- diphenyl tetrazolium bromide assay] MTT assay was performed. To determine whether shRNA interference (shRNAi) is capable of sensitizing B16F10 cells to chemotherapeutic agents, cells were transfected with an LD50 of each of the recombinant plasmids, treated with varying concentrations of doxorubicin or cisplatin 24 h post-transfection, and analyzed 48 h later for inhibition of cell proliferation using the MTT assay. We observed that WT1-RNAi and the two chemotherapeutic agents acted synergistically to inhibit B16F10 cell proliferation. The greatest inhibition of cell proliferation was observed with the WT1-2/cisplatin (91%) and WT1-1/cisplatin combinations (85%). WT1 silencing using shRNAi induced the chemosensitization of cells to doxorubicin and cisplatin, with the greatest inhibition (85%) of cell proliferation being observed in the cells treated with the WT1-2/cisplatin 6 ng/µl combination. Our results provide direct evidence that WT1 gene silencing has a synergistic effect with chemotherapeutic drugs and sensitizes B16F10 melanoma cells to doxorubicin and cisplatin. This suggests that these combination strategies are potentially utilized in melanoma therapy.
Collapse
Affiliation(s)
- Pablo Zapata-Benavides
- Laboratorio de Inmunología y Virología, Departamento de Microbiología e Inmunología, Facultad de Ciencias Biológicas de la Universidad Autónoma de Nuevo León, San Nicolás de los Garza, N.L. México
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Zhao Y, Zhang S, Cui S, Wang B, Zhang S. Peptide-based cationic liposome-mediated gene delivery. Expert Opin Drug Deliv 2011; 9:127-39. [DOI: 10.1517/17425247.2011.630387] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
15
|
Morandi F, Corrias MV, Levreri I, Scaruffi P, Raffaghello L, Carlini B, Bocca P, Prigione I, Stigliani S, Amoroso L, Ferrone S, Pistoia V. Serum levels of cytoplasmic melanoma-associated antigen at diagnosis may predict clinical relapse in neuroblastoma patients. Cancer Immunol Immunother 2011; 60:1485-95. [PMID: 21660451 DOI: 10.1007/s00262-011-1052-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Accepted: 05/24/2011] [Indexed: 12/13/2022]
Abstract
The high molecular weight melanoma-associated antigen (HMW-MAA) and the cytoplasmic melanoma-associated antigen (cyt-MAA/LGALS3BP) are expressed in melanoma. Their serum levels are increased in melanoma patients and correlate with clinical outcome. We investigated whether these molecules can serve as prognostic markers for neuroblastoma (NB) patients. Expression of cyt-MAA and HMW-MAA was evaluated by flow cytometry in NB cell lines, patients' neuroblasts ((FI)-NB), and short-term cultures of these latter cells (cNB). LGALS3BP gene expression was evaluated by RT-qPCR on (FI)-NB, cNB, and primary tumor specimens. Soluble HMW-MAA and cyt-MAA were tested by ELISA. Cyt-MAA and HMW-MAA were expressed in NB cell lines, cNB, and (FI)-NB samples. LGALS3BP gene expression was higher in primary tumors and cNB than in (FI)-NB samples. Soluble cyt-MAA, but not HMW-MAA, was detected in NB cell lines and cNBs supernatants. NB patients' serum levels of both antigens were higher than those of the healthy children. High cyt-MAA serum levels at diagnosis associated with higher incidence of relapse, independently from other known risk factors. In conclusion, both HMW-MAA and cyt-MAA antigens, and LGALS3BP gene, were expressed by NB cell lines and patients' neuroblasts, and both antigens' serum levels were increased in NB patients. Elevated serum levels of cyt-MAA at diagnosis correlated with relapse, supporting that cyt-MAA may serve as early serological biomarker to individuate patients at higher risk of relapse that may require a more careful follow-up, after being validated in a larger cohort of patients at different time-points during follow-up. Given its immunogenicity, cyt-MAA may also be a potential target for NB immunotherapy.
Collapse
Affiliation(s)
- Fabio Morandi
- Laboratory of Oncology, G. Gaslini Children's Hospital, Largo G. Gaslini 5, 16148 Genoa, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Pisano M, Pagnan G, Dettori MA, Cossu S, Caffa I, Sassu I, Emionite L, Fabbri D, Cilli M, Pastorino F, Palmieri G, Delogu G, Ponzoni M, Rozzo C. Enhanced anti-tumor activity of a new curcumin-related compound against melanoma and neuroblastoma cells. Mol Cancer 2010; 9:137. [PMID: 20525240 PMCID: PMC2898702 DOI: 10.1186/1476-4598-9-137] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Accepted: 06/03/2010] [Indexed: 12/15/2022] Open
Abstract
Background Sharing the common neuroectodermal origin, melanoma and neuroblastoma are tumors widely diffused among adult and children, respectively. Clinical prognosis of aggressive neuroectodermal cancers remains dismal, therefore the search for novel therapies against such tumors is warranted. Curcumin is a phytochemical compound widely studied for its antioxidant, anti-inflammatory and anti-cancer properties. Recently, we have synthesized and tested in vitro various curcumin-related compounds in order to select new anti-tumor agents displaying stronger and selective growth inhibition activity on neuroectodermal tumors. Results In this work, we have demonstrated that the new α,β-unsaturated ketone D6 was more effective in inhibiting tumor cells growth when compared to curcumin. Normal fibroblasts proliferation was not affected by this treatment. Clonogenic assay showed a significant dose-dependent reduction in both melanoma and neuroblastoma colony formation only after D6 treatment. TUNEL assay, Annexin-V staining, caspases activation and PARP cleavage unveiled the ability of D6 to cause tumor cell death by triggering apoptosis, similarly to curcumin, but with a stronger and quicker extent. These apoptotic features appear to be associated with loss of mitochondrial membrane potential and cytochrome c release. In vivo anti-tumor activity of curcumin and D6 was surveyed using sub-cutaneous melanoma and orthotopic neuroblastoma xenograft models. D6 treated mice exhibited significantly reduced tumor growth compared to both control and curcumin treated ones (Melanoma: D6 vs control: P < 0.001 and D6 vs curcumin P < 0.01; Neuroblastoma: D6 vs both control and curcumin: P < 0.001). Conclusions Our data indicate D6 as a good candidate to develop new therapies against neural crest-derived tumors.
Collapse
Affiliation(s)
- Marina Pisano
- Istituto di Chimica Biomolecolare, CNR, Sassari, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Chao M, Jiawei X, Zhongxin J, Kuang A. Anionic long‐circulating liposomes for delivery of radioiodinated antisense oligonucleotides. EUR J LIPID SCI TECH 2010. [DOI: 10.1002/ejlt.200900283] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Ma Chao
- Department of Nuclear Medicine, Affiliated Hospital, Qingdao University, Qingdao, Shandong Province, China
| | - Xie Jiawei
- Department of Stomatology, Affiliated Hospital, Qingdao University, Qingdao, Shandong Province, China
| | - Jiang Zhongxin
- Department of Chemical Examination, Affiliated Hospital, Qingdao University, Qingdao, Shandong Province, China
| | - Anren Kuang
- Department of Nuclear Medicine, Affiliated Hospital, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
18
|
Santos AO, da Silva LCG, Bimbo LM, de Lima MCP, Simões S, Moreira JN. Design of peptide-targeted liposomes containing nucleic acids. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2009; 1798:433-41. [PMID: 20004174 DOI: 10.1016/j.bbamem.2009.12.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2009] [Revised: 11/08/2009] [Accepted: 12/01/2009] [Indexed: 11/15/2022]
Abstract
Anticancer systemic gene silencing therapy has been so far limited by the inexistence of adequate carrier systems that ultimately provide an efficient intracellular delivery into target tumor cells. In this respect, one promising strategy involves the covalent attachment of internalizing-targeting ligands at the extremity of PEG chains grafted onto liposomes. Therefore, the present work aims at designing targeted liposomes containing nucleic acids, with small size, high encapsulation efficiency and able to be actively internalized by SCLC cells, using a hexapeptide (antagonist G) as a targeting ligand. For this purpose, the effect of the liposomal preparation method, loading material (ODN versus siRNA) and peptide-coupling procedure (direct coupling versus post-insertion) on each of the above-mentioned parameters was assessed. Post-insertion of DSPE-PEG-antagonist G conjugates into preformed liposomes herein named as stabilized lipid particles, resulted in targeted vesicles with a mean size of about 130 nm, encapsulation efficiency close to 100%, and a loading capacity of approximately 5 nmol siRNA/mumol of total lipid. In addition, the developed targeted vesicles showed increased internalization in SCLC cells, as well as in other tumor cells and HMEC-1 microvascular endothelial cells. The improved cellular association, however, did not correlate with enhanced downregulation of the target protein (Bcl-2) in SCLC cells. These results indicate that additional improvements need to be performed in the future, namely by ameliorating the access of the nucleic acids to the cytoplasm of the tumor cells following receptor-mediated endocytosis.
Collapse
Affiliation(s)
- Adriana O Santos
- Laboratory of Pharmaceutical Technology, University of Coimbra, Portugal
| | | | | | | | | | | |
Collapse
|
19
|
Sequential administration with oxaliplatin-containing PEG-coated cationic liposomes promotes a significant delivery of subsequent dose into murine solid tumor. J Control Release 2009; 142:167-73. [PMID: 19861140 DOI: 10.1016/j.jconrel.2009.10.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2009] [Revised: 09/27/2009] [Accepted: 10/19/2009] [Indexed: 11/22/2022]
Abstract
Recently, we designed a PEG-coated cationic liposome to achieve dual targeting delivery of l-OHP to both tumor endothelial cells and tumor cells in a solid tumor. The targeted liposomal l-OHP formulation showed an efficient antitumor activity in a murine tumor model after three sequential liposomal l-OHP injections. This led us to assume that prior dosing with liposomes might enhance the intra-tumoral accumulation of a subsequent dose, and hence improve the therapeutic efficacy of entrapped l-OHP. The present study shows that while a single liposomal l-OHP injection does not enhance tumor accumulation of subsequent test-PEG-coated cationic liposomes, two sequential injections of liposomal l-OHP do. Cumulative cytotoxic effects of l-OHP delivered by PEG-coated cationic liposomes led to deep diffusion of a subsequent dose of liposomal l-OHP in solid tumor presumably as a result of the enlarged intra-tumoral interstitial space. Our study suggests that sequential injections of a targeted liposomal anticancer drug is of significant clinical and practical importance in enhancing the delivery of adequate quantities of anticancer agents into intractable solid tumors, and thereby may achieve a significant anticancer efficacy.
Collapse
|
20
|
Tang Y, McGoron AJ. Combined effects of laser-ICG photothermotherapy and doxorubicin chemotherapy on ovarian cancer cells. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2009; 97:138-44. [PMID: 19811928 DOI: 10.1016/j.jphotobiol.2009.09.001] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2008] [Revised: 08/31/2009] [Accepted: 09/03/2009] [Indexed: 12/31/2022]
Abstract
Doxorubicin (DOX) is an anthracycline antibiotic widely used in cancer chemotherapy. Its use is limited by cardiac toxicity and drug resistance. Hyperthermia can aid the functionality of DOX, but current hyperthermia delivery methods are hard to apply selectively and locally. The slow temperature increase associated with the external heating may lead to thermal tolerance in cancer cells. The FDA approved dye indocynine green (ICG) has been demonstrated to absorb near-infrared (NIR) light at 808 nm (ideal for tissue penetration) and emit the energy as heat, making it an ideal agent for localized hyperthermia with a rapid rate of temperature increase. The purpose of this study was to investigate the in vitro cytotoxic effect of combined chemotherapy and hyperthermia to a DOX resistant ovarian cancer cell line (SKOV-3). The effect of two different heating methods, ICG induced rapid rate heating and an incubator induced slow rate heating, were compared. All the experiments were conducted in 96-well plates. Cells were subjected to different concentrations of DOX and 60 min 43 degrees C incubation or 5 microM of ICG with 1 min 808 nm NIR laser. SRB assay was used to measure cell proliferation. ICG itself without laser irradiation was not toxic to SKOV-3 cells. The two types of hyperthermia individually produced similar cytotoxicity. DOX by itself was toxic with an IC(50) value of about 5 microM. Hyperthermia in combination with DOX achieved significantly greater cell killing/growth inhibition at all DOX concentrations compared to DOX alone. A subadditive cytotoxic effect was observed by combining DOX and 60 min 43 degrees C incubation which lead to a lowered DOX IC(50) value of about 1 microM. This value was even lower with 1 min laser-ICG photothermotherapy (0.1 microM) and, though not statistically significant, a synergistic effect may exist between DOX and laser-ICG photothermotherapy. The rate of heating may have an effect on chemotherapy-hyperthermia interaction. In conclusion, the combination of photothermal therapy and chemotherapy may provide a valuable tool for cancer treatment with minimized side effect.
Collapse
Affiliation(s)
- Yuan Tang
- Department of Biomedical Engineering, Florida International University, 10555 West Flagler Street, Miami, FL 33174, USA
| | | |
Collapse
|
21
|
Obata Y, Saito S, Takeda N, Takeoka S. Plasmid DNA-encapsulating liposomes: Effect of a spacer between the cationic head group and hydrophobic moieties of the lipids on gene expression efficiency. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2009; 1788:1148-58. [DOI: 10.1016/j.bbamem.2009.02.014] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2008] [Revised: 01/19/2009] [Accepted: 02/12/2009] [Indexed: 11/16/2022]
|
22
|
Pastorino F, Di Paolo D, Piccardi F, Nico B, Ribatti D, Daga A, Baio G, Neumaier CE, Brignole C, Loi M, Marimpietri D, Pagnan G, Cilli M, Lepekhin EA, Garde SV, Longhi R, Corti A, Allen TM, Wu JJ, Ponzoni M. Enhanced Antitumor Efficacy of Clinical-Grade Vasculature-Targeted Liposomal Doxorubicin. Clin Cancer Res 2008; 14:7320-9. [DOI: 10.1158/1078-0432.ccr-08-0804] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Purpose: In vivo evaluation of good manufacturing practice-grade targeted liposomal doxorubicin (TVT-DOX), bound to a CD13 isoform expressed on the vasculature of solid tumors, in human tumor xenografts of neuroblastoma, ovarian cancer, and lung cancer.
Experimental Design: Mice were implanted with lung, ovarian, or neuroblastoma tumor cells via the pulmonary, peritoneal, or orthotopic (adrenal gland) routes, respectively, and treated, at different days post inoculation, with multiple doses of doxorubicin, administered either free or encapsulated in untargeted liposomes (Caelyx) or in TVT-DOX. The effect of TVT-DOX treatment on tumor cell proliferation, viability, apoptosis, and angiogenesis was studied by immunohistochemical analyses of neoplastic tissues and using the chick embryo chorioallantoic membrane assay.
Results: Compared with the three control groups (no doxorubicin, free doxorubicin, or Caelyx), statistically significant improvements in survival was seen in all three animal models following treatment with 5 mg/kg (maximum tolerated dose) of TVT-DOX, with long-term survivors occurring in the neuroblastoma group; increased survival was also seen at a dose of 1.7 mg/kg in mice bearing neuroblastoma or ovarian cancer. Minimal residual disease after surgical removal of neuroblastoma primary mass, and the enhanced response to TVT-DOX, was visualized and quantified by bioluminescence imaging and with magnetic resonance imaging. When treated with TVT-DOX, compared with Caelyx, all three tumor models, as assayed by immunohistochemistry and chorioallantoic membrane, showed statistically significant reductions in cell proliferation, blood vessel density, and microvessel area, showing increased cell apoptosis.
Conclusion: TVT-DOX should be evaluated as a novel angiostatic strategy for adjuvant therapy of solid tumors.
Collapse
Affiliation(s)
- Fabio Pastorino
- 1Experimental Therapies Unit, Laboratory of Oncology, G. Gaslini Children's Hospital
| | - Daniela Di Paolo
- 1Experimental Therapies Unit, Laboratory of Oncology, G. Gaslini Children's Hospital
| | | | - Beatrice Nico
- 5Department of Human Anatomy, University of Bari, Bari, Italy
| | | | | | - Gabriella Baio
- 4Department of Radiology, National Cancer Institute, Genoa, Italy
| | | | - Chiara Brignole
- 1Experimental Therapies Unit, Laboratory of Oncology, G. Gaslini Children's Hospital
| | - Monica Loi
- 1Experimental Therapies Unit, Laboratory of Oncology, G. Gaslini Children's Hospital
| | - Danilo Marimpietri
- 1Experimental Therapies Unit, Laboratory of Oncology, G. Gaslini Children's Hospital
| | - Gabriella Pagnan
- 1Experimental Therapies Unit, Laboratory of Oncology, G. Gaslini Children's Hospital
| | | | | | | | - Renato Longhi
- 7Istituto di Chimica del Riconoscimento Molecolare, Consiglio Nazionale delle Ricerche
| | - Angelo Corti
- 8Immunobiotechnology Unit, San Raffaele Institute, Milan, Italy; and
| | - Theresa M. Allen
- 9Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Jinzi J. Wu
- 6Ambrilia Biopharma Inc., Verdun, Quebec, Canada
| | - Mirco Ponzoni
- 1Experimental Therapies Unit, Laboratory of Oncology, G. Gaslini Children's Hospital
| |
Collapse
|
23
|
Leaman DW. Recent progress in oligonucleotide therapeutics: antisense to aptamers. Expert Opin Drug Discov 2008; 3:997-1009. [DOI: 10.1517/17460441.3.9.997] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|