Minireviews Open Access
Copyright ©The Author(s) 2021. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Oncol. Apr 15, 2021; 13(4): 231-237
Published online Apr 15, 2021. doi: 10.4251/wjgo.v13.i4.231
Nanotechnology and pancreatic cancer management: State of the art and further perspectives
Damiano Caputo, Tommaso Farolfi, Roberto Passa, Roberto Coppola, Department of General Surgery, University Campus Bio-Medico di Roma, Rome 00128, Italy
Daniela Pozzi, Giulio Caracciolo, Department of Molecular Medicine, Sapienza University of Rome, Rome 00161, Italy
ORCID number: Damiano Caputo (0000-0001-7058-1945); Daniela Pozzi (0000-0001-6158-5880); Tommaso Farolfi (0000-0003-1932-2846); Roberto Passa (0000-0002-1119-4643); Roberto Coppola (0000-0001-5798-0714); Giulio Caracciolo (0000-0002-8636-4475).
Author contributions: Caputo D contributed to conception or design of the work; Farolfi T and Passa R contributed to data collection; Caputo D, Pozzi D and Caracciolo G contributed to data analysis and interpretation; Caputo D and Caracciolo G contributed drafting the article; Caputo D, Pozzi D, Coppola R and Caracciolo G contributed to critical revision of the article; all authors did the final approval of the version to be published.
Conflict-of-interest statement: All Authors declare that they do not have any conflict of interest.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Damiano Caputo, FACS, MD, Associate Professor, Surgeon, Department of General Surgery, University Campus Bio-Medico di Roma, Via Álvaro del Portillo, 200, Rome 00128, Italy. d.caputo@unicampus.it
Received: January 12, 2021
Peer-review started: January 12, 2021
First decision: February 11, 2021
Revised: February 21, 2021
Accepted: March 12, 2021
Article in press: March 12, 2021
Published online: April 15, 2021

Abstract

Pancreatic ductal adenocarcinoma (PDAC) represents a leading cause of cancer death and is often diagnosticated too late to allow adequate treatments. Lots of biomarkers have been discovered in lasts years but, to date, there is a lack of low-cost and non-invasive tools for PDAC early detection. Nonetheless, drugs commonly used in PDAC treatment do not allow achieving long-term satisfying results. Nanotechnology is gaining importance in both PDAC early detection and treatment. The main implications of nanotechnology in cancer diagnosis lay in the ability that nanoparticles have on concentrate the alteration in human proteome caused by cancer. Nanoparticle-enabled blood tests have been demonstrated to reach high rate of sensitivity (up to 85%) and specificity (up to 100%). In the field of cancer therapy nanoparticles can be used as nanocarriers able to reach specific tumour’s cells and selectively release the drug they contain into them. A literature review was carried out with the aim to assess the state of the art and highlight the future perspectives of nanotechnology in PDAC early detection and therapy.

Key Words: Pancreatic ductal adenocarcinoma, Biomarkers, Nanotechnology, Nanoparticles, Protein corona, Early detection

Core Tip: Pancreatic ductal adenocarcinoma (PDAC) is very lethal. Surgery is the only effective treatment. Unfortunately, only 20% of PDACs can be radically resected because of the lack of tools for early diagnosis and since chemotherapy agents fail to reach satisfying prognosis. Nanoparticles can amplify the cancer-induced proteome alteration allowing early cancer detection and can also be used in cancer treatment as carriers improving the effectiveness of chemotherapy. This review focuses the actual and futures implications of nanotechnology in PDAC diagnosis and treatment.



INTRODUCTION

Pancreatic ductal adenocarcinoma (PDAC) represents a sneaky disease that often runs asymptomatic in its early stage. Symptoms as intense pain or jaundice that led to medical consult frequently occur when the tumour is already in an advanced stage. Therefore, PDAC first diagnosis is generally reached when the local invasion or distant metastases are present. This behaviour, in association with the biology of the tumour, account for the poor prognosis that affect the majority of PDAC patients[1].

Surgery may improve patient’s prognosis; regrettably, radical resection is precluded to almost 80% of the affected because only a minority of PDACs are diagnosed early enough[2].

Chemotherapy, alone or in association with radiotherapy is commonly used in neoadjuvant or adjuvant settings or in presence of metastatic disease. Unfortunately, to date, PDAC responsivity to these treatments remains poor[3].

To date, there is a lack of cheap, user-friendly and non-invasive tools for PDAC early detection. Food and Drug Administration (FDA) approved only carbohydrate antigen (Ca) 19.9 for use in daily clinical practice; however, the American Society of Clinical Oncology does not recommend it in a diagnostic phase because of its poor sensitivity and specificity[4].

Over the last decade, a large number of different biomarkers have been identified; many proved their efficacy, alone or in combination with Ca 19.9, in detecting PDAC and/or distinguish PDAC from other pancreatic benign diseases (e.g., chronic pancreatitis)[5].

Unfortunately, almost all of them failed to become reproducible in routinely practice since resulting from complex, expensive and laborious technology (e.g., genetic sequencing, transcriptomic expression profiling, proteomic and metabolomic profiling) and not meeting the ASSURED (Affordable, Sensitive, Specific, User-friendly, Rapid and robust, Equipment-free and Deliverable to end-users) criteria requested by the World Health Organization (WHO)[6].

In the field of cancer treatment, immunotherapy represents the “newer kid on the block”. This science is based on the enhancement of the immune system activity against cancer cells. Despite being promising, immunotherapy still has its “dark side” when considered for PDAC. PDAC microenvironment has been identified as a leading cause of failure of this therapeutic strategy[7].

On these bases, it is clear that both newer PDAC early diagnosis strategies and more efficient therapies represent the challenges to win and hot topics of research.

Since nanomedicine is gaining momentum in cancer management, aim of the present work was to assess the state of the art and to highlight the future perspectives of nanotechnology in PDAC early detection and therapy.

METHODS

To tackle the above-mentioned issue, PubMed database was queried for articles published up to December 2020. Terms pancreatic cancer, pancreatic adenocarcinoma, nanotechnology, nanomedicine, nanoparticles, diagnosis, detection, therapy and management have been used in different combinations. References reported in the selected articles have been also considered as other bibliographic sources. Only articles published in English with available full text and without limitation concerning article types (original articles, review, etc.), subjects involved in the research (e.g., humans, mice), experimental condition (in vitro or in vivo) and population size have been considered.

NANOTECHNOLOGY AND PDAC EARLY DIAGNOSIS

Nanotechnology is a multidisciplinary scientific field involving physics, engineering, chemistry and biology. This science is based on the use of objects whose dimensions are in the nanoscale (range 1 nm–100 nm) (http://data.europa.eu/eli/reco/2011/696/oj.): the nanoparticles (NPs). In recent years, this science is gaining more and more momentum opening interesting scenarios in the management of different types of tumours.

When NPs interact with organic fluids (e.g., plasma), they are covered by a shell of biomolecules, forming the so-called biomolecular corona (BC). Since proteins represent the most abundant elements forming the BC, this structure is often called Protein Corona (PC) too.

The shape of PC depends on NPs’ physiochemical properties (e.g. materials, size, electric charge, hydrophobicity)[8], environmental factors (temperature, incubation time)[9] and experimental source characteristics (e.g., plasma, urine, human, murine)[10].

Actually, since NPs are able to concentrate even poorly abundant plasma proteins, characterization of PC may reveal changes in protein concentration that cannot be detected by common laboratory tests.

The main implications of nanotechnology in cancer diagnosis and treatment lay in the ability that NPs have to concentrate cancer-induced alteration in human proteome. Nevertheless, it has been proved that different patients affected by different pathological conditions have personalized PC[11].

On this basis and considering that proteins alterations are the hallmark of cancerogenesis, NP-enabled tools (NETs) have been investigated for early cancer detection. Moreover, by the use of common techniques (e.g., light scattering, electrophoresis) nanotechnology proved to fully meet the above-mentioned WHO criteria.

Prostate cancer has been the first tumour investigated for early detection based on the development of NETs[12]. Further similar tests have been lately developed and applied to other solid neoplasms[13].

In 2014, a pilot study was conducted analysing plasma samples collected from 11 patients affected by untreated PDAC and 13 patients undergoing surgery for benign disease (“healthy controls”). The samples were let to interact with anionic liposomes (ALs) made of 1,2-dioleoyl-sn-glycero-3-phospho-(1'-rac-glycerol) (DOPG); the obtained PCs general characteristics (i.e., dimension, surface charge and protein composition) and their protein abundances were assessed by Dynamic Light Scattering and Laser Doppler Electrophoresis techniques. The Authors demonstrated significant differences in the surface charge (the zeta potential) and size between PCs formed after interaction of NPs with plasma of PDAC and healthy (i.e., non-oncological) subjects. To explain these findings, different protein patterns constituting PCs of cancer patients and controls were hypothesized. Indeed, PCs of PDAC patients were significantly more enriched than those of healthy subjects. This enrichment was particularly due to a subset of plasma proteins with molecular weight comprised between 30 and 60 kDa. The lack of proteomics-based mass spectrometry investigations did not allow to fully characterize the proteins forming the PCs in the two groups[14].

Moreover, during the last recent years, different NPs have been tested in order to improve the accuracy of NETs in detecting PDAC. Among them, particular relevance gained graphene oxide (GO) nanoflakes. Differently from other NPs, GO nanoflakes bind low levels of the most abundant proteins present in human blood (e.g., albumin) and have the ability to adsorb other poorly-concentrated plasma proteins. As shown by Papi et al[15], when PCs formed around GO nanoflakes of 25 pancreatic ductal adenocarcinoma patients were compared with those of 25 controls, PDAC patients PCs were significantly less enriched of proteins with molecular weight ranging between 20-30 kDa. This difference allowed distinguishing between cancer and non-cancer subjects with a sensitivity of 92%.

Nevertheless, the studies performed analyzing the PC of PDACs demonstrated that also the stage of the disease determines changes in PC composition[16]. Particularly, SDS-PAGE analysis of PCs formed around 1,2-dioleoyl-sn-glycero-3-phospho-(1'-rac-glycerol) liposomes allowed to distinguish patients affected by T1-T2 PDACs from T3s and metastatic ones. More in detail, PC intensity profiles of T1-T2 pancreatic ductal adenocarcinomas significantly differed from those of the T3s because of the dissimilar abundance of a subset of proteins with molecular weight ranging between 25 and 50 kDa (p 0.04). Comparing T1-T2 to metastatic PDACs, significant differences in intensity profiles were also found in the subgroups of proteins with molecular weight of 25-50 kDa (p 0.01) and 50-120 kDa (p 0.04). The differences highlighted in the protein intensities would reflect the stage-related different expression of proteins (e.g., VEGF, CDK2, adhesion molecules, etc.) already reported as result of more sophisticated and expensive technologies[14].

All the studies focused on PDAC early diagnosis and considered in this paper have been summarized in Table 1.

Table 1 Nanotechnology and pancreatic ductal adenocarcinoma early diagnosis.
Year
Ref.
NP type
Main findings
2012Walkey et al[8]Gold, Silica, Polystyrene Characteristics of NPs (material, size, electric charge, hydrophobicity) influence the composition of the PC
2012Monopoli et al[10]Silica, Carbon, Iron oxide, PolystyreneDifferent experimental sources (e.g., plasma, urine), affect the composition of the PC
2013Tenzer et al[9]Silica, PolystyreneEnvironmental factors (e.g., incubation time) influence the composition of PC
2014Caracciolo et al[14]Liposomes After interaction of NPs with plasma of PDACs and non-oncological subjects, differences in the electric charge (zeta potential) and size of the PCs allow to distinguish between the two groups
2015Zheng et al[12]GoldGold-NPs successfully used for early detection of prostate cancer
2017Corbo et al[11]Silica, Polystyrene, Gold, LiposomesDifferent patients affected by different pathological conditions have “personalized” PC
2018Caputo et al[16]LiposomesAnalysis of PCs formed around DOPG liposomes allowed to distinguish patients affected by T1-T2 PDACs from T3s and metastatic ones
2019Künzel et al[13]Silica Successful application of silica NPs in early diagnosis of the head and neck solid squamous carcinoma
2019Papi et al[15]GOAnalysis of BC formed around GO nanoflakes distinguished PDAC from healthy subjects (sensitivity 92%)
NANOTECHNOLOGY AND PDAC THERAPY

Due to the modest results commonly achieved by conventional drugs, nanotechnologies have been tested to improve the effectiveness of chemotherapy agents by increasing the availability of the drug in the tumour and decreasing the toxicity reducing its effects on host cells[17].

Essentially, NPs can be loaded with drugs and used as nanocarriers able to reach the tumour and selectively release the drug they contain into it.

Many NPs as liposomes, micelles, dendrimers, exomes, organic and inorganic NPs and different mechanisms of action as endocytosis, fusion with plasma membranes, active or passive targeting have been studied and increasingly used for solid cancer treatment[18].

In 2008, Patra et al[19] demonstrated, in vitro and in vivo, a significant inhibition of pancreatic cancer cells proliferation and growth using a formulation of gold NPs transporting cetuximab (C225) and gemcitabine.

In 2015, liposomal irinotecan (Onivyde®) was approved by the FDA for clinical use in metastatic PDAC, who already have received gemcitabine. The efficacy of Onivyde’s® was demonstrated in a randomized trial performed on a series of 417 patients affected by metastatic PDACs. Patients who received the combination of Onivyde® and 5-fluorouracil (5-FU)/Leucovorin gained on average 2 mo of survival and showed an average delay in the time to tumor growth of 3.1 mo when compared to those who received only 5-FU/Leucovorin[20].

However, it has been demonstrated that these liposomal formulations lose their selectivity over the time, due to the exposure to blood proteins, and their toxic effect increases. As shown by Mirshafiee and colleagues, after exposure to plasma, NPs present significant inhibition (up to 94%) of their targeting ability. This inhibition is due to the formation of the PC that impairs the biodistribution and the cellular uptake of the NPs, and to the effect of the organic filters (liver and spleen)[21].

Nonetheless, Liu demonstrated in PDAC animal models that, when compared with liposomes, mesoporous silica nanoparticles (MSNs) improve irinotecan loading and efficacy[22]. More in detail, the Author developed a supported lipid bilayer coated MSN carrier with increased loading capacity and cancer cells killing ability when compared to the conventional liposomal formulation. Furthermore, these nanocarriers showed improved stability decreasing the irinotecan toxicity by the reduction of the systemic leakage of the drug. The MSN carrier developed by Liu and coworkers was also more efficient in the treatment of pancreatic cancer metastatic cells and, according to the Author opinion, could be used in FOLFIRINOX regimen treatment for PDAC.

NP albumin-bound paclitaxel, better known in clinical practice as nab-paclitaxel or abraxane, is the newest drug approved for PDAC treatment and is widely used in association with gemcitabine in gem-abraxane combined therapeutic settings[23].

As demonstrated in mice by Meng et al[24] in 2015, coated bilayer MSNs can also improve the synergic delivery of gemcitabine and paclitaxel in human PDACs.

In this paper, Meng also focused on the role that MSN nanocarriers have in overcoming the challenges of PDAC stromal barrier and abnormal perfusion that have been widely recognized among the main factors contributing to the failure of oncological treatments.

On the basis of the interesting results obtained using MSNs, these NPs have been tested also in the immunotherapy for PDAC. MSNs loaded with oxaliplatin and indoximod, an inhibitor of immunosuppressive indoleamine 2,3-dioxygenase, showed their ability in improving innate and adaptative immunity against PDAC in vaccination approaches, systemic and local administration[25]. The Authors also reported the innovation of using a nanocarrier to produce a synergistic immune response by the concomitant delivery of an immunogenic cell death stimulus and interfering in immune suppression.

Nevertheless, nanocarriers have been proposed for different gene therapies for PDAC. In this field, NPs can be used to improve the biological availability and selectivity of nucleic acids as small interfering RNA (siRNA) or microRNA and reduce the immune system activity against these therapeutic agents[26].

Despite the large number and the quality of advances in NP-based therapies for PDAC, the research for a safe, non-toxic stable, biocompatible and biodegradable nanovector is still the gap to fill. In this field, chitosan may pave the way for the development of efficient and not toxic drug delivery system[27]. As reported in the review of Sadoughi et al[27], chitosan proved its efficacy in PDAC therapy as nanocarrier of both gemcitabine and 5-FU.

All the studies focused on the application of nanotechnology in PDAC therapy considered in this paper have been summarized in Table 2.

Table 2 Nanotechnology and pancreatic ductal adenocarcinoma therapy.
Year
Ref.
NP type
Main Findings
2008Patra et al[19]GoldGold NPs used as nanocarrier improve the effect of cetuximab and gemcitabine in reducing the growth and the proliferation of pancreatic cancer cells in murine model.
2013Mirshafiee et al[21]SilicaThe exposure to human plasma significantly reduces the targeting ability of NPs.
2015Meng et al[24]MSNCoated bilayer MSNs improve the synergic delivery of gemcitabine and paclitaxel in PANC-1 injected in murine models.
2015FDA[20]Liposomal irinotecan (Onivyde®)Approval of Onivyde® for clinical use in metastatic PDAC, who already have received gemcitabine.
2016Liu et al[22]MSNIn KPC pancreatic cancer cells injected into murine models, biocompatibility and therapeutic efficacy of MSNs-irinotecan formulation are superior to liposomal-irinotecan formulation.
2017Lu et al[25]MSNMSNs improve oxaliplatin and indoximod bioavailability in KPC pancreatic cancer cells injected into murine models.
2017Shi et al[23]NP albuminFDA approved the clinical use of NP albumin-bound paclitaxel (Abraxane) for pancreatic cancer.
2020Sadoughi et al[27]ChitosanThe use of chitosan as carrier of gemcitabine and 5-FU improves therapeutic effect decreasing toxicity in pancreatic cancer cells.
CONCLUSION

According to this literature review and taking into account that potential bias due to possible missing analyzed articles cannot be excluded, NP-based tools seem to be promising in PDAC management.

If further confirmed on larger series, results obtained from original and pioneering experiences may lead to the development of fast and cheap tool that could be used for early PDAC detection.

Nonetheless, if PDAC tumour dimension and presence of metastases will be confirmed relevant in NP-enabled blood test accuracy, nanotechnology could also be used together with standard clinical and pathological staging system; this “molecular” staging may provide useful information for PDAC prognosis and therapy.

Nanotechnologies have already demonstrated their usefulness in improving the therapeutic response to pancreatic cancer. However, future research efforts in this area must on the one hand be aimed at improving nanotechnology interfaces and, on the other hand, answer the unresolved questions relating to the bioavailability and clearance of nanovectors in order to increase efficacy and reduce toxic effect.

A more accurate understanding of the interactions between NPs and biological fluids represents the prerequisite for ensuring that nanomedicines confirm to be a valid potential weapon against this lethal disease.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Medicine, research and experimental

Country/Territory of origin: Italy

Peer-review report’s scientific quality classification

Grade A (Excellent): A

Grade B (Very good): B, B

Grade C (Good): C, C

Grade D (Fair): D

Grade E (Poor): 0

P-Reviewer: Hao Y, Uhlmann D, Wang XB, Yip-Schneider M S-Editor: Zhang H L-Editor: A P-Editor: Li JH

References
1.  He XY, Yuan YZ. Advances in pancreatic cancer research: moving towards early detection. World J Gastroenterol. 2014;20:11241-11248.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Yeo TP, Hruban RH, Leach SD, Wilentz RE, Sohn TA, Kern SE, Iacobuzio-Donahue CA, Maitra A, Goggins M, Canto MI, Abrams RA, Laheru D, Jaffee EM, Hidalgo M, Yeo CJ. Pancreatic cancer. Curr Probl Cancer. 2002;26:176-275.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Adamska A, Domenichini A, Falasca M. Pancreatic Ductal Adenocarcinoma: Current and Evolving Therapies. Int J Mol Sci. 2017;18.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Locker GY, Hamilton S, Harris J, Jessup JM, Kemeny N, Macdonald JS, Somerfield MR, Hayes DF, Bast RC Jr;  ASCO. ASCO 2006 update of recommendations for the use of tumor markers in gastrointestinal cancer. J Clin Oncol. 2006;24:5313-5327.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Jimenez-Luna C, Torres C, Ortiz R, Dieguez C, Martinez-Galan J, Melguizo C, Prados JC, Caba O. Proteomic biomarkers in body fluids associated with pancreatic cancer. Oncotarget. 2018;9:16573-16587.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Kosack CS, Page AL, Klatser PR. A guide to aid the selection of diagnostic tests. Bull World Health Organ. 2017;95:639-645.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Mucciolo G, Roux C, Scagliotti A, Brugiapaglia S, Novelli F, Cappello P. The dark side of immunotherapy: pancreatic cancer. Cancer Drug Resist. 2020;3:491-520.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Walkey CD, Chan WC. Understanding and controlling the interaction of nanomaterials with proteins in a physiological environment. Chem Soc Rev. 2012;41:2780-2799.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Tenzer S, Docter D, Kuharev J, Musyanovych A, Fetz V, Hecht R, Schlenk F, Fischer D, Kiouptsi K, Reinhardt C, Landfester K, Schild H, Maskos M, Knauer SK, Stauber RH. Rapid formation of plasma protein corona critically affects nanoparticle pathophysiology. Nat Nanotechnol. 2013;8:772-781.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Monopoli MP, Aberg C, Salvati A, Dawson KA. Biomolecular coronas provide the biological identity of nanosized materials. Nat Nanotechnol. 2012;7:779-786.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Corbo C, Molinaro R, Tabatabaei M, Farokhzad OC, Mahmoudi M. Personalized protein corona on nanoparticles and its clinical implications. Biomater Sci. 2017;5:378-387.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Zheng T, Pierre-Pierre N, Yan X, Huo Q, Almodovar AJ, Valerio F, Rivera-Ramirez I, Griffith E, Decker DD, Chen S, Zhu N. Gold nanoparticle-enabled blood test for early stage cancer detection and risk assessment. ACS Appl Mater Interfaces. 2015;7:6819-6827.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Künzel J, Gribko A, Lu Q, Stauber RH, Wünsch D. Nanomedical detection and downstream analysis of circulating tumor cells in head and neck patients. Biol Chem. 2019;400:1465-1479.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Caracciolo G, Caputo D, Pozzi D, Colapicchioni V, Coppola R. Size and charge of nanoparticles following incubation with human plasma of healthy and pancreatic cancer patients. Colloids Surf B Biointerfaces. 2014;123:673-678.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Papi M, Palmieri V, Digiacomo L, Giulimondi F, Palchetti S, Ciasca G, Perini G, Caputo D, Cartillone MC, Cascone C, Coppola R, Capriotti AL, Laganà A, Pozzi D, Caracciolo G. Converting the personalized biomolecular corona of graphene oxide nanoflakes into a high-throughput diagnostic test for early cancer detection. Nanoscale. 2019;11:15339-15346.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Caputo D, Cartillone M, Cascone C, Pozzi D, Digiacomo L, Palchetti S, Caracciolo G, Coppola R. Improving the accuracy of pancreatic cancer clinical staging by exploitation of nanoparticle-blood interactions: A pilot study. Pancreatology. 2018;18:661-665.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Din FU, Aman W, Ullah I, Qureshi OS, Mustapha O, Shafique S, Zeb A. Effective use of nanocarriers as drug delivery systems for the treatment of selected tumors. Int J Nanomedicine. 2017;12:7291-7309.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Wei QY, Xu YM, Lau ATY. Recent Progress of Nanocarrier-Based Therapy for Solid Malignancies. Cancers (Basel). 2020;12.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Patra CR, Bhattacharya R, Wang E, Katarya A, Lau JS, Dutta S, Muders M, Wang S, Buhrow SA, Safgren SL, Yaszemski MJ, Reid JM, Ames MM, Mukherjee P, Mukhopadhyay D. Targeted delivery of gemcitabine to pancreatic adenocarcinoma using cetuximab as a targeting agent. Cancer Res. 2008;68:1970-1978.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  FDA. FDA Approves Onivyde Combo Regimen for Advanced Pancreatic Cancer. Oncol Times. 2015;37:8.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Mirshafiee V, Mahmoudi M, Lou K, Cheng J, Kraft ML. Protein corona significantly reduces active targeting yield. Chem Commun (Camb). 2013;49:2557-2559.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Liu X, Situ A, Kang Y, Villabroza KR, Liao Y, Chang CH, Donahue T, Nel AE, Meng H. Irinotecan Delivery by Lipid-Coated Mesoporous Silica Nanoparticles Shows Improved Efficacy and Safety over Liposomes for Pancreatic Cancer. ACS Nano. 2016;10:2702-2715.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Shi J, Kantoff PW, Wooster R, Farokhzad OC. Cancer nanomedicine: progress, challenges and opportunities. Nat Rev Cancer. 2017;17:20-37.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Meng H, Wang M, Liu H, Liu X, Situ A, Wu B, Ji Z, Chang CH, Nel AE. Use of a lipid-coated mesoporous silica nanoparticle platform for synergistic gemcitabine and paclitaxel delivery to human pancreatic cancer in mice. ACS Nano. 2015;9:3540-3557.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Lu J, Liu X, Liao YP, Salazar F, Sun B, Jiang W, Chang CH, Jiang J, Wang X, Wu AM, Meng H, Nel AE. Nano-enabled pancreas cancer immunotherapy using immunogenic cell death and reversing immunosuppression. Nat Commun. 2017;8:1811.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Kurtanich T, Roos N, Wang G, Yang J, Wang A, Chung EJ. Pancreatic Cancer Gene Therapy Delivered by Nanoparticles. SLAS Technol. 2019;24:151-160.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Sadoughi F, Mansournia MA, Mirhashemi SM. The potential role of chitosan-based nanoparticles as drug delivery systems in pancreatic cancer. IUBMB Life. 2020;72:872-883.  [PubMed]  [DOI]  [Cited in This Article: ]