2Changshu Second People’s Hospital, Changshu 215500, Jiangsu Province, China
3Zhenjiang Integrative Medicine Hospital, Zhenjiang 212001, Jiangsu Province, China
Keywords: Pancreatic Cancer; Liposomes; Nanoparticles; Therapeutic Drug Delivery
The study shows that the develop high-content gemcitabine PEGylated liposomes to reverse gemcitabine resistance in pancreatic tumor cells. The mechanism of drug loading into liposomes was also investigated. Remote loading was not appropriate for filling gemcitabine into liposomes. pKa > 4.6 for basic drugs and intra-liposomal precipitation of loaded compounds were recommended as an extra requirement to the recent criteria for remote loading using ammonium sulfate gradient (pKa < 11). High DL is essential for liposomes to reverse gemcitabine resistance in pancreatic cell lines [25]. Increased interstitial fluid pressure, decrease access to the nanomedicine medications to cancer [26]. Furthermore, pancreatic cancer restricts the penetration of drugs to tumor cells due to its extracellular matrix [27]. Passive targeting does not stop nanocarriers accumulation in other body parts such us liver and spleen due to their fenestrated endothelium [28]. Therefore, the production of nanomedicine drugs next generation is certainly warranted with advanced functions. Nanomedicine Second generation medications are assembled on a drug-delivery system with dynamic targeting nano-carriers with stimulatory response properties. Therefore, the second generation of nano-drugs enhance the targeting efficacy [29]. The next generation has formed many nanomedicines at the preclinical level, and some of these have been tested clinically, but have not been accepted for commercial use.
This review shows that anti-CA199 cys-diabody providing exact molecular imaging in cancer xenograft models demonstrates target binding of pancreatic tumor cells with distributing good treatment to the target site [12,37]. Improvement in the five years survival rate of the pancreatic ductal adenocarcinoma (PDAC) quickly required. Though combination four drugs, FOLFIRINOX (5-fluorouracil, leucovorin, oxaliplatin, comprising and irinotecan), provides a healthier survival result than the recently used drug gemcitabine, the previous treatment regimen has a very toxic effect and stopped its using in patients with decent result. Meanwhile, Irinotecan adds toxic effect to FOLFIRINOX like gastrointestinal and bone marrow toxicity, in order to decrease the toxic effect of this medication through the design of mesoporous silica nanoparticle (MSNP), can by covering the liposomes with a high dose of proton gradient filler for irinotecan. The effective steadiness of lipid bilayer coated mesoporous silica nanoparticle (LB-MSNP) carrier causes fewer medicine distributions systemically and more absorptions at cancer targeted sites. The lipid bilayer coated mesoporous silica nanoparticle nanocarrier was more effective for the treating of metastatic cancer. Decrease leakage and slowly release of the medication by lipid bilayer coated mesoporous silica nanoparticle carrier impressively decreased the rate of the toxic effect on gastrointestinal, liver and bone marrow compared to a liposomal carrier system. We suggest that combination of the high effective rate and decreased toxic effect by the LB-MSNP carrier may help in the use of Irinotecan in the pancreatic ductal adenocarcinoma as first-line therapeutic agent to increase its survival rate [38].
The ability to distribute cells under in vivo and in vitro conditions, liposomes with antitumor activity conjugated to HA of different molecular weights, the first primary ligand for CD44. We demonstrated that HA-conjugated liposomes received higher cell-to-CD44 expression in pancreatic adenocarcinoma than unconjugated liposomes [12].
The TSLs also exist limitations, (1) their capacity of targeting is weak. It is difficult to avoid the role of the mononuclear phagocytic system (2) the heating time is too long can also cause damage to normal tissue.
The important task of increasing antitumor efficacy is to increase drug accumulation in cancerous tissues and also to control the size. The fluorescence of INPs presented actual imaging monitor for subcellular locating and metabolic distribution in vivo. Near-infrared imaging in photo-thermal therapy and vivo demonstrated that 68 nm INPs displayed the toughest effectiveness to destroy cancer growth due to plentiful buildup in BxPC-3 xenograft tumor model. These results show that a nontoxic, size-dependent, theranostic INPs model was assembled for in vivo tumor imaging and photothermic therapy without any side effect [67].
The researchers recognized that chemical connection of the anti-cancer medication doxorubicin on to squalene, a normal lipid originator of the cholesterol’s biosynthesis, directed to the development of squalenoyl doxorubicin (SQ-Dox) nanoassemblies of 130-nm, with a unique “loop-train” arrangement. Cell culture viability tests and apoptosis assays indicated that SQ-Dox nano-assemblies showed similar anti-proliferative and cytotoxicity than the natural doxorubicin because of its action to stimulate apoptotic mediators, these are caspase-3 and polymerase (ADP-ribose). Concerning toxic effect, SQDox nanoscale assembly presents a five-fold higher maximum tolerated dose than free drugs, and furthermore, the cardiac profile of SQ-Dox nano-assemblies demonstrated that the SQ-Dox nano-assemblies didn’t cause myocardial problems and lesions, which are those who persuaded by the unrestricted doxorubicin usage. If use together, these results prove that the SQ-Dox nanoassemblies mark cancer cells extra sensitive to doxorubicin and decrease the myocardial toxic effect, therefore presenting a extraordinary improvement in the medication’s therapeutic index [68].
Injection of Gemcitabine liposome (stealth liposomes) has simplified the targeting of gemcitabine for the tumor treatment. We mainly review drug delivery systems based on liposomes, which can increase pharmacokinetics, decrease toxic effects and possibly improve tumor uptake, for the treatment pancreatic tumors. In this study, since extrusion technology was used to make a sterile preparation of liposomes, the process included aseptic production process and sterile filtration. During the preparation, it has been found that the lipid concentration,emulsification speed and time, the homogenization times and pattern, the lipid solution temperature are all critical parameters for the character of the gemcitabine liposome injection. The particle size method and zeta potential method to characterize a PEGylated liposomal drug formulation of anti-tumor agent gemcitabine developed. The methods are specific, precise, reproducible and sensitive, therefore they are suitable for determination of particle size and zeta potential of gemcitabine liposome injection. Negative staining technology of transmission electron microscopy revealed that gemcitabine liposome injection has a typical morphology, which enables liposomal surfaces could be seen so additional visual information on the stealth liposome can be routinely obtained in a fast and reliable manner. Moreover, the above three methods are simple, fast and would be used for continuous quality control of gemcitabine liposome injection when it moves to cGMP production scale [69].
Liposomes are vesicles composed of single or multiple phospholipid bilayers that can be full with a different type of content including genetic materials and chemotherapeutic drugs. Liposomes can improve drug solubility and stability, are biodegradable, and exhibit low toxicity. They have already proven to be viable clinically, many of them FDA-approved liposome formulations in existence for the treatment of cancer. Liposomes have been used for in vitro gene transfection to protect antisense oligonucleotides, siRNA, and shRNA from degradation and improve transfection efficiency and targeting. For example, liposomal delivery of both pancreatic and duodenal homeobox 1 (PDX-1) and ZIP4-targeted shRNA was shown to prevent tumor progression in immune-deficient mice. By adding of surface ligands can help to liposomes to target cells of interest, thereby helping to decrease the toxic effect of the therapeutic agents and enrich concentrations in target tissues. Liposomes can also be PEGylated to increase stability and prolong the circulation time of a drug. In a study by Cosco and colleagues, PEGylated liposomes loaded with gemcitabine increased survival and reduced tumor progression and toxic effect in severely compromised immune-deficient (SCID) xenograft mouse models of pancreatic malignancy compared to controls treated with standard gemcitabine. Improved liposomal delivery efficiency may further allow lower concentrations of drugs to be used for the same effect, more circulation time, improve medication internalization, and cut unwanted toxicity.
Ultra-small superparamagnetic iron oxides (USPIOs) and doxorubicin (DOX) was ready by the transient binding and reverse-phase evaporation technique, and it was also conjugated with the anti-mesothelin monoclonal antibody by post-insertion process to target anti-mesothelin-over expressed cells of the pancreatic tumor. In vivo and in vitro properties of anti-mesothelin antibody-conjugated PEGylated liposomal DOX and USPIOs (M-PLDU and PEGylated nano Immune liposome without any antibody conjugation [PLDU]) were assessed both human tumor of pancreatic cell line Panc-1 cell and in the cancer of pancreas xenograft animal model. The results of this study showed that M-PLDU controlled advanced inhibitory effect on the growth of the cancer and distribution assay of tissue further more verified that M-PLDUs might selectively collect in the tumor xenograft. This study shows that M-PLDU not only well retains the inherent MRI capability of USPIO, but also significantly enhances the targeted delivery of USPIO and therapeutic agents in pancreatic tumor cells [70].
Gold nanorods (GNR) with longitudinal surfaces are adjustable between 600 and 1100 nm. GNR is conjugated to transferrin (TF) to target pancreatic tumor cells. Two-photon imaging of bioconjugated GNR-evoked receptor-mediated bioconjugate absorption into Panc-1 cells, over expression of transferrin receptor (TFR). Bioconjugated GNR formulations cause very little toxic effects, indicating that they are biocompatible and potentially suitable for targeting two-photon bioimaging [83].
Other investigational therapeutic agents include monoclonal antibodies against targets which is EGFR, vascular endothelial growth factor receptor (VEGFR), mucin 1 (MUC1), and mesothelin. Antibody therapies have the potential to inhibit tumor growth and angiogenesis and provide a method of treating otherwise resistant cancers. Oncolytic viral therapies, often using herpes simplex virus and adenovirus, are engineered to selectively replicate in tumor cells, targeting the cells for lysis, cell-to-cell fusion, or immune response.
Active drug |
Product name |
Indications |
Daunorubicin |
DaunoXome |
Kaposi’s sarcoma |
Doxurubicin |
Mycet |
Combinational therapy of recurrent breast cancer |
Doxorubicin in PEG-liposomes |
Doxil/Caelyx |
Refractory Kaposi’s sarcoma; ovarian cancer; recurrent breast cancer |
Cytarabine |
DepoCyt |
Lymphomatous meningitis |
Amphotericin B |
AmBisome |
Fungal infections |
Vincristine |
Onco TCS |
Non-Hodgkin’s lymphoma |
Nystatin |
Nyotran |
Topical antifungal agent |
Lurtotecan |
NX211 |
Ovarian cancer |
All-trans retinoic acid |
Altragen |
Acute promyelocytic leukaemia; non-Hodgkin’s lymphoma; renalcell |
Platinum compounds |
Platar |
Solid tumours |
Annamycin |
|
Doxorubicin-resistant tumours |
E1A gene |
Various tumours |
|
DNA plasmid encoding HLA-B7 and α2 microglobulin |
Allovectin-7 |
Metastatic melanoma |
Liposomes for various drugs and diagnostic agents (lipoMASC |
|
Broad applications |
Recent advancements in the development of therapeutic agents and distribution modalities for better-quality targeting, efficacy, and clinical outcomes in pancreatic cancer are extremely promising. However, because the data are mostly preclinical and the clinical trials are few, more research is necessary to further refine these methods, confirm their safety, and improve delivery specificity. Even if targeted delivery is accomplished and the drug enters the cell, it is possible that there will be a response failure. Therefore, a greater understanding of pancreatic cell biology is also crucial since the presence of multiple mutations may make single-target treatments insufficient. Emerging targeted therapeutic approaches may be exciting, viable opportunities that will tangibly improve the management of this devastating disease. In the past decade, studies have shown that nanoparticles used in a number pancreatic tumor have a great advantage. However, the targeted liposomes in the treatment of pancreatic tumor in the conveyor are a very complicated process. Therefore, the need to develop new nanoparticles to progress the therapeutic effect of the drug, and decrease systemic toxicity. Most studies in experimental animals and future research is needed to broaden the research and study of the human body model.
- Jemal A, Murray T, Ward E, Samuels A, Tiwari RC, Ghafoor A, et al. Cancer statistics, 2005. CA Cancer J Clin. 2005;55(1):10-30.
- Berger AC, Meszoely IM, Ross EA, Watson JC, Hoffman JP. Undetectable preoperative levels of serum CA 19-9 correlate with improved survival for patients with resectable pancreatic adenocarcinoma. Ann Surg Oncol. 2004;11(7):644-649.
- Ferrone CR, Brennan MF, Gonen M, Coit DG, Fong Y, Chung S, et al. Pancreatic adenocarcinoma: the actual 5-year survivors. J Gastrointest Surg. 2008;12(4):701-706.
- Mahalingam D, Giles F. Challenges in developing targeted therapy for pancreatic adenocarcinoma. Expert Opin Ther Targets. 2008;12(11):1389-1401. doi: 10.1517/14728222.12.11.1389
- Fujii T. Extended lymphadenectomy in pancreatic cancer is crucial. World J Surg. 2013;37(8):1778-1781. doi: 10.1007/s00268-013-2039-0
- Wray CJ, Ahmad SA, Matthews JB, Lowy AM. Surgery for pancreatic cancer: recent controversies and current practice. Gastroenterology. 2005;128(6):1626-1641.
- Yang F, Jin C, Yang D, Jiang Y, Li J, Di Y, et al. Magnetic functionalised carbon nanotubes as drug vehicles for cancer lymph node metastasis treatment. Eur J Cancer. 2011;47(12):1873-1882. doi: 10.1016/j.ejca.2011.03.018
- Alexiou C, Schmid RJ, Jurgons R, et al. Targeting cancer cells: magnetic nanoparticles as drug carriers. European Biophysics Journal. 2006;35(5):446-450.
- Chen J, Wang L, Yao Q, Ling R, Li K, Wang H. Drug concentrations in axillary lymph nodes after lymphatic chemotherapy on patients with breast cancer. Breast Cancer Research. 2004;6(4):1.
- Madani SY, Naderi N, Dissanayake O, Tan A, Seifalian AM. A new era of cancer treatment: carbon nanotubes as drug delivery tools. Int J Nanomedicine. 2011;6:2963-2979. doi: 10.2147/IJN.S16923
- Yu X, Zhang Y, Chen C, Yao Q, Li M. Targeted drug delivery in pancreatic cancer. Biochimica et Biophysica Acta (BBA)-Reviews on Cancer. 2010;1805(1):97-104.
- McCarroll J, Teo J, Boyer C, Goldstein D, Kavallaris M, Phillips P. Potential applications of nanotechnology for the diagnosis and treatment of pancreatic cancer. Front Physiol. 2014;5:2. doi: 10.3389/fphys.2014.00002
- Braganza JM, Hunt LP, Warwick F. Relationship between pancreatic exocrine function and ductal morphology in chronic pancreatitis. Gastroenterology. 1982;82(6):1341-1347.
- Torchilin V. Antibody-modified liposomes for cancer chemotherapy. Expert Opin Drug Deliv. 2008;5(9):1003-1025.
- Muthu MS, Singh S. Targeted nanomedicines: effective treatment modalities for cancer, AIDS and brain disorders. Nanomedicine. 2009;4(1):105-118. doi: 10.2217/17435889.4.1.105
- Torchilin VP. Recent advances with liposomes as pharmaceutical carriers. Nat Rev Drug Discov. 2005;4(2):145-160.
- Matsumura Y, Maeda H. A new concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs. Cancer research. 1986;46(12 Part 1):6387-6392.
- Maeda H. The enhanced permeability and retention (EPR) effect in tumor vasculature: the key role of tumor-selective macromolecular drug targeting. Adv Enzyme Regul. 2001;41(1):189-207.
- Maeda H, Nakamura H, Fang J. The EPR effect for macromolecular drug delivery to solid tumors: Improvement of tumor uptake, lowering of systemic toxicity, and distinct tumor imaging in vivo. Advanced drug delivery reviews. 2013;65(1):71-79.
- Maeda H, Wu J, Sawa T, Matsumura Y, Hori K. Tumor vascular permeability and the EPR effect in macromolecular therapeutics: a review. Journal of controlled release. 2000;65(1-2):271-284.
- Nichols JW, Bae YH. EPR: evidence and fallacy. Journal of Controlled Release. 2014;190:451-464.
- Wang X, Yang L, Chen ZG, Shin DM. Application of nanotechnology in cancer therapy and imaging. CA: a cancer journal for clinicians. 2008;58(2):97-110. doi: 10.3322/CA.2007.0003
- Haley B, Frenkel E. Nanoparticles for drug delivery in cancer treatment. Urol Oncol. 2008;26(1):57-64. doi: 10.1016/j.urolonc
- Asokan A, Schaffer DV, Samulski RJ. The AAV vector toolkit: poised at the clinical crossroads. Molecular Therapy. 2012;20(4):699-708.
- Xu H, Paxton J, Lim J, et al. Development of high-content gemcitabine pegylated liposomes and their cytotoxicity on drug-resistant pancreatic tumour cells. Pharmaceutical research. 2014;31(10):2583-2592. doi: 10.1007/s11095-014-1353-z
- Jain RK, Stylianopoulos T. Delivering nanomedicine to solid tumors. Nature reviews Clinical oncology. 2010;7(11):653-664. doi: 10.1038/nrclinonc.2010.139
- Provenzano PP, Cuevas C, Chang AE, Goel VK, Von Hoff DD, Hingorani SR. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer cell. 2012;21(3):418-429. doi: 10.1016/j.ccr.2012.01.007
- Gaumet M, Vargas A, Gurny R, Delie F. Nanoparticles for drug delivery: the need for precision in reporting particle size parameters. European journal of pharmaceutics and biopharmaceutics. 2008;69(1):1-9.
- Farokhzad OC, Langer R. Nanomedicine: developing smarter therapeutic and diagnostic modalities. Advanced drug delivery reviews. 2006;58(14):1456-1459.
- Adams GP, Schier R, McCall AM, et al. High affinity restricts the localization and tumor penetration of single-chain fv antibody molecules. Cancer research. 2001;61(12):4750-4755.
- Gosk S, Moos T, Gottstein C, Bendas G. VCAM-1 directed immunoliposomes selectively target tumor vasculature in vivo. Biochim Biophys Acta. 2008;1778(4):854-863. doi: 10.1016/j.bbamem.2007.12.021
- Dicheva BM, ten Hagen TL, Schipper D, Seynhaeve AL, van Rhoon GC, Eggermont AM, et al. Targeted and heat-triggered doxorubicin delivery to tumors by dual targeted cationic thermosensitive liposomes. Journal of controlled release. 2014;195:37-48. doi: 10.1016/j.jconrel.2014.07.058
- Sudimack J, Lee RJ. Targeted drug delivery via the folate receptor. Advanced drug delivery reviews. 2000;41(2):147-162.
- Hogrefe RI, Lebedev AV, Zon G, Pirollo KF, Rait A, Zhou Q, et al. Chemically modified short interfering hybrids (siHYBRIDS): nanoimmunoliposome delivery in vitro and in vivo for RNAi of HER-2. Nucleosides Nucleotides Nucleic Acids. 2006;25(8):889-907.
- Pirollo KF, Rait A, Zhou Q, Hwang SH, Dagata JA, Zon G, et al. Materializing the potential of small interfering RNA via a tumor-targeting nanodelivery system. Cancer research. 2007;67(7):2938-2943.
- Pirollo KF, Zon G, Rait A, Zhou Q, Yu W, Hogrefe R, et al. Tumor-targeting nanoimmunoliposome complex for short interfering RNA delivery. Human gene therapy. 2006;17(1):117-124.
- Bunt T, Malone JM, Moody M, Davidson J, Karpman R. Frequency of vascular injury with blunt trauma-induced extremity injury. Am J Surg. 1990;160(2):226-228.
- Liu X, Situ A, Kang Y, Yanan Kang, Katie Rose Villabroza, Yupei Liao, et al. Irinotecan delivery by lipid-coated mesoporous silica nanoparticles shows improved efficacy and safety over liposomes for pancreatic cancer. ACS nano. 2016;10(2):2702-2715.
- Gaucher G, Marchessault RH, Leroux JC. Polyester-based micelles and nanoparticles for the parenteral delivery of taxanes. Journal of controlled release. 2010;143(1):2-12. doi: 10.1016/j.jconrel.2009.11.012
- Gong J, Chen M, Zheng Y, Wang S, Wang Y. Polymeric micelles drug delivery system in oncology. J Control Release. 2012;159(3):312-323. doi: 10.1016/j.jconrel.2011.12.012
- Nicolas J, Mura S, Brambilla D, Mackiewicz N, Couvreur P. Design, functionalization strategies and biomedical applications of targeted biodegradable/biocompatible polymer-based nanocarriers for drug delivery. Chem Soc Rev. 2013;42(3):1147-1235. doi: 10.1039/c2cs35265f
- Peer D, Karp JM, Hong S, Farokhzad OC, Margalit R, Langer R. Nanocarriers as an emerging platform for cancer therapy. Nature nanotechnology. 2007;2(12):751-760. doi:10.1038/nnano.2007.387
- Deng C, Jiang Y, Cheng R, Meng F, Zhong Z. Biodegradable polymeric micelles for targeted and controlled anticancer drug delivery: promises, progress and prospects. Nano Today. 2012;7(5):467-480.
- Owen SC, Chan DP, Shoichet MS. Polymeric micelle stability. Nano Today. 2012;7(1):53-65.
- O'Reilly RK, Hawker CJ, Wooley KL. Cross-linked block copolymer micelles: functional nanostructures of great potential and versatility. Chemical Society Reviews. 2006;35(11):1068-1083.
- Read ES, Armes SP. Recent advances in shell cross-linked micelles. Chemical Communications. 2007(29):3021-3035.
- van Nostrum CF. Covalently cross-linked amphiphilic block copolymer micelles. Soft Matter. 2011;7(7):3246-3259.
- Daniels TR, Delgado T, Rodriguez JA, Helguera G, Penichet ML. The transferrin receptor part I: Biology and targeting with cytotoxic antibodies for the treatment of cancer. Clinical immunology. 2006;121(2):144-158.
- Cheng Y, Zak O, Aisen P, Harrison SC, Walz T. Structure of the human transferrin receptor-transferrin complex. Cell. 2004;116(4):565-576.
- Fuchs H, Lücken U, Tauber R, Engel A, Geßner R. Structural model of phospholipid-reconstituted human transferrin receptor derived by electron microscopy. Structure. 1998;6(10):1235-1243.
- Lebrón JA, Bennett MJ, Vaughn DE, Chirino AJ, Snow PM, Mintier GA, et al. Crystal structure of the hemochromatosis protein HFE and characterization of its interaction with transferrin receptor. Cell. 1998;93(1):111-123.
- Ciechanover A, Schwartz AL, Lodish HF. Sorting and recycling of cell surface receptors and endocytosed ligands: the asialoglycoprotein and transferrin receptors. J Cell Biochem. 1983;23(1‐4):107-130.
- Hémadi M, Kahn PH, Miquel G, El Hage Chahine J-M. Transferrin's mechanism of interaction with receptor 1. Biochemistry. 2004;43(6):1736-1745.
- Das GA, Shah VI. Correlation of transferrin receptor expression with histologic grade and immunophenotype in chronic lymphocytic leukemia and non-Hodgkin's lymphoma. Hematologic pathology. 1989;4(1):37-41.
- Habeshaw J, Lister T, Stansfeld A, Greaves M. Correlation of transferrin receptor expression with histological class and outcome in non-Hodgkin lymphoma. The Lancet. 1983;1(8323):498-501.
- Kondo K, Noguchi M, Mukai K, et al. Transferrin receptor expression in adenocarcinoma of the lung as a histopathologic indicator of prognosis. Chest. 1990;97(6):1367-1371.
- Prior R, Reifenberger G, Wechsler W. Transferrin receptor expression in tumours of the human nervous system: relation to tumour type, grading and tumour growth fraction. Virchows Arch A Pathol Anat Histopathol. 1990;416(6):491-496.
- Seymour G, Walsh M, Lavin M, Strutton G, Gardiner R. Transferrin receptor expression by human bladder transitional cell carcinomas. Urological research. 1987;15(6):341-344.
- Yang DC, Wang F, Elliott RL, Head JF. Expression of transferrin receptor and ferritin H-chain mRNA are associated with clinical and histopathological prognostic indicators in breast cancer. Anticancer Res. 2000;21(1B):541-549.
- White S, Taetle R, Seligman PA, Rutherford M, Trowbridge IS. Combinations of anti-transferrin receptor monoclonal antibodies inhibit human tumor cell growth in vitro and in vivo: evidence for synergistic antiproliferative effects. Cancer Res. 1990;50(19):6295-6301.
- Callens C, Moura I, Lepelletier Y, et al. Recent advances in adult T-cell leukemia therapy: focus on a new anti-transferrin receptor monoclonal antibody. Leukemia. 2008;22(1):42-48.
- Ng PP, Cruz JSD, Sorour DN, Stinebaugh JM, Shin SU, Shin DS, et al. An anti-transferrin receptor-avidin fusion protein exhibits both strong proapoptotic activity and the ability to deliver various molecules into cancer cells. Proc Natl Acad Sci U S A. 2002;99(16):10706-10711.
- Ng PP, Helguera G, Daniels TR, Lomas SZ, Rodriguez JA, Schiller G, et al. Molecular events contributing to cell death in malignant human hematopoietic cells elicited by an IgG3-avidin fusion protein targeting the transferrin receptor. Blood. 2006;108(8):2745-2754.
- Arpicco S, Lerda C, Dalla Pozza E, Costanzo C, Tsapis N, Stella B, et al. Hyaluronic acid-coated liposomes for active targeting of gemcitabine. Eur J Pharm Biopharm. 2013;85(3):373-380. doi: 10.1016/j.ejpb.2013.06.003
- Viudez A, Ramirez N, Hernandez-Garcia I, Carvalho F, Vera R, Hidalgo M. Nab-paclitaxel: a flattering facelift. Crit Rev Oncol Hematol. 2014;92(3):166-180. doi: 10.1016/j.critrevonc.2014.06.001
- Ta T, Bartolak-Suki E, Park E-J, Karrobi K, McDannold NJ, Porter TM. Localized delivery of doxorubicin in vivo from polymer-modified thermosensitive liposomes with MR-guided focused ultrasound-mediated heating. Journal of Controlled Release. 2014;194:71-81. doi: 10.1016/j.jconrel.2014.08.013
- Zhao P, Zheng M, Yue C, Luo Z, Gong P, Gao G, et al. Improving drug accumulation and photothermal efficacy in tumor depending on size of ICG loaded lipid-polymer nanoparticles. Biomaterials. 2014;35(23):6037-6046. doi: 10.1016/j.biomaterials.2014.04.019
- Maksimenko A, Dosio F, Mougin J, Ferrero A, Wack S, Reddy LH, et al. A unique squalenoylated and nonpegylated doxorubicin nanomedicine with systemic long-circulating properties and anticancer activity. Proc Natl Acad Sci U S A. 2014;111(2):E217-E226. doi: 10.1073/pnas.1313459110
- Zhou Q, Liu L, Zhang D, Fan X. Preparation and characterization of gemcitabine liposome injections. Pharmazie. 2012;67(10):844-847.
- Deng L, Ke X, He Z, Yang D, Gong H, Zhang Y, et al. A MSLN-targeted multifunctional nanoimmunoliposome for MRI and targeting therapy in pancreatic cancer. Int J Nanomedicine. 2012;7:5053-5065. doi: 10.2147/IJN.S34801
- Patra CR, Bhattacharya R, Mukhopadhyay D, Mukherjee P. Application of gold nanoparticles for targeted therapy in cancer. Journal of Biomedical Nanotechnology. 2008;4(2):99-132.
- Webster TJ. Nanomedicine: what’s in a definition? International journal of nanomedicine. 2006;1(2):115.
- Zhang L, Gu F, Chan J, Wang A, Langer R, Farokhzad O. Nanoparticles in medicine: therapeutic applications and developments. Clin Pharmacol Ther. 2008;83(5):761-769.
- Cuenca AG, Jiang H, Hochwald SN, Delano M, Cance WG, Grobmyer SR. Emerging implications of nanotechnology on cancer diagnostics and therapeutics. Cancer. 2006;107(3):459-466.
- Gdowski A, Ranjan AP, Mukerjee A, Vishwanatha JK. Nanobiosensors: role in cancer detection and diagnosis. Infectious Diseases and Nanomedicine. 2014:33-58.
- Marchal F, Pic E, Pons T, Dubertret B, Bolotine L, Guillemin F. Quantum dots in oncological surgery: the future for surgical margin status. Bull Cancer. 2008;95(12):1149-1153. doi: 10.1684/bdc.2008.0757
- Doucas H, Garcea G, Neal C, Manson M, Berry D. Chemoprevention of pancreatic cancer: a review of the molecular pathways involved, and evidence for the potential for chemoprevention. Pancreatology. 2006;6(5):429-439.
- Efthimiou E, Crnogorac-Jurcevic T, Lemoine N. Pancreatic cancer genetics. Pancreatology. 2001;1(6):571-575.
- Jaffee EM, Hruban RH, Canto M, Kern SE. Focus on pancreas cancer. Cancer cell. 2002;2(1):25-28.
- Maitra A, Hruban RH. Pancreatic cancer. Annual review of pathology. 2008;3:157-188.
- Mimeault M, Brand RE, Sasson AA, Batra SK. Recent advances on the molecular mechanisms involved in pancreatic cancer progression and therapies. Pancreas. 2005;31(4):301-316.
- Patra CR, Bhattacharya R, Mukhopadhyay D, Mukherjee P. Fabrication of gold nanoparticles for targeted therapy in pancreatic cancer. Advanced drug delivery reviews. 2010;62(3):346-361. doi: 10.1016/j.addr.2009.11.007
- Zhu J, Yong K-T, Roy I, Hu R, Ding H, Zhao L, et al. Additive controlled synthesis of gold nanorods (GNRs) for two-photon luminescence imaging of cancer cells. Nanotechnology. 2010;21(28):285106. doi: 10.1088/0957-4484/21/28/285106