Abstract
Purpose of review
Organ transplantation is a life-saving procedure and the only option for patients with end-organ failure. Immune therapeutics have been key to the success of organ transplantation. However, immune therapeutics are still unable to eliminate graft rejection and their toxicity has been implicated in poorer long-term transplant outcomes. Targeted nanodelivery has the potential to enhance not only the therapeutic index but also the bioavailability of the immune therapeutics. One of the key sites of immune therapeutics delivery is lymph node where the priming of immune cells occur. The focus of this review is on nanomedicine research to develop the targeted delivery of immune therapeutics to lymph nodes for controlling immune activation.
Recent findings
As nanomedicine creates its niche in clinical care, it provides novel immunotherapy platforms for transplant recipients. Draining lymph nodes are the primary loci of immune activation and represent a formidable site for delivery of wide variety of immune therapeutics. There have been relentless efforts to improve the properties of nanomedicines, to have in-depth knowledge of antigen and drug loading, and, finally, to explore various routes of passive and active targeted delivery to lymph nodes.
Summary
The application of nanotechnology principles in the delivery of immune therapeutics to the lymph node has created enormous excitement as a paradigm shifting approach that enables targeted delivery of a gamut of molecules to achieve a desired immune response. Therefore, innovative strategies that improve their efficacy while reducing their toxicity are among the highest unmet needs in transplantation.
Keywords: drug delivery, nanomedicine, nanoparticles, transplantation
INTRODUCTION
Application of nanomedicine in transplantation
Organ transplantation is the optimal therapy for patients with irreversible, end-stage organ disease. Although immune therapeutics have improved over time, a large number of transplant patients, however, suffer from the consequences of allograft rejection. Furthermore, immune therapeutics are implicated in the pathogenesis of organ failure (i.e. calcineurin inhibitor toxicity). Contributing to an increased incidence of post-transplant diabetes, hyperlipidemia, and also playing pathogenesis of accelerated cardiovascular disease, which is the leading cause of death in the transplanted population [1-7]. One of the major clinical approaches to improve long-term transplant outcome is to improve the therapeutic index of immune therapeutics (increasing the efficacy but reducing the toxicity) [1, 5, 8-14]. Therefore, there have been many conventional approaches of sparing, minimizing, or withdrawing calcineurin inhibitor. However, these strategies have limited challenges such as unmodified toxicity, development of new sets of toxicity caused by replacing the drug, as well as higher rejection rates when immunosuppression was reduced [3, 15-17].
Nanotechnology is a multidisciplinary field involving design and engineering of submicron structures. Application of nanotechnology in medicine has rapidly evolved over the last two decades. There are numerous reports on introduction of nanotechnology into different aspects of medicine including diagnosis (biological detection, sensors, and imaging), treatment (drug delivery and tissue engineering), monitoring, prediction, and prevention of diseases [18-21].
Nanocarriers have been developed to treat a wide variety of diseases, mainly because of their specific features that distinguish them from other modalities for instance antibody–drug conjugates [22]. Liposomes, which are nanoparticles composed of a lipidic bilayer enclosing an aqueous core (incorporating drugs, nucleic acids, etc.), were the first nanocarriers to be approved, making them the oldest nanotherapeutic platform [23]. Polymer–drug conjugates, colloidal nanocarriers wherein drug molecules are covalently attached to the polymeric backbone, and polymeric micelles composed of diblock polymers encapsulating drugs, are other examples of nanotherapies that followed clinical approval [23]. These nanoparticles represent the majority of clinically validated and approved nanomedicines [24]. Polymeric nanoparticles, protein-based nanoparticles, and dendrimers are also nanoparticles that are undergoing clinical translation. Other types of nanomaterials used for clinical nanodiagnostics and nanotherapeutic applications include viral nanoparticles, inorganic nanoparticles composed of gold, silica, iron oxide, and hafnium oxide, in addition to oncolytic viruses, which are under clinical investigation [25]. The majority of these nanocarriers are used for oncology applications, as they, in particular, become trapped within tumors through the so-called ‘enhanced permeation and retention’ (EPR) effect [26]. The discovery of the EPR effect in the 1980s suggested that colloidal nanoparticles can accumulate in tumors due to fenestrations in tumor vessel endothelium, as a result of excessive branching, and chaotic vasculature that caused the breakdown of tight junctions and disruption of the basement membrane [27, 28]. Hence, oncology applications of nanomedicines have been widely studied, resulting in major interest in the design of nanoparticles of different sizes, shapes, and surface characteristics aiming to improve tumor or target-site accumulation [29].
Advantages of nanomedicines include, first, ability to deliver hydrophobic drugs; second, improved pharmacokinetics and biodistribution of therapeutics [30]; third, enhanced bioavailabity of small therapeutics and biological drugs [22, 31, 32]; fourth, improved tolerability and therapeutic indices of drugs by reducing marginal side-effect and systemic toxicity of the therapeutics by selective delivery to the target site; fifth, delivering therapeutics across biological barriers, for instance, epithelial and endothelial barriers [33-36]; sixth, conjugation of multiple targeting moieties to design nanocarriers with considerably enhanced selectivity; seventh, loading multiple therapeutics into the nanocarriers, another unique advantage of these delivery platforms, without affecting pharmacokinetics of the individual drugs; and eighth, sustained and controlled drug release, which is another well desired feature of nanocarriers [37]. For many applications, notably immune therapeutics drug delivery, slow extended release of the therapeutics is extremely essential to improve therapeutic index.
Nano-sized drug carriers
As mentioned earlier, polymeric nanoparticles and liposomes are two of the most well studied drug nanocarriers. Liposomes are one of the first types of nanocarriers mainly investigated for delivery of hydrophobic drugs. Loading of the drug into a single or double lipid layer increases the solubility and alters the biodistribution pattern of the drug. The size and surface chemistry of the liposomes dictates the biodistribution and not the drug molecules. Yet, liposomes suffer from burst release of the drug, ineffectiveness in intracellular delivery of drug molecules and evading biobarriers [38-41].
Among the polymers developed to formulate polymeric nanoparticles, poly(lactic-co-glycolic acid) (PLGA) has gained considerable research interests because of its favorable properties including biodegradability, biocompatibility, and approval of the Food and Drug Administration as well as European Medicine Agency[42]. PLGA nanoparticles reduce the chance of therapeutic degradation and improve bioavailability of poorly water-soluble therapeutic agents, and facilitate controlled release [32, 43-45].
Biodistribution of nanoparticles, and effect of size, shape, and surface charge
A better understanding of obstacles and barriers nanoparticles face upon administration into the bloodstream has led to the design of nanoparticles with optimized physiochemical properties, prolonged circulation time, organ-specific accumulation, reduced toxicity, and enhanced therapeutic index [46]. Very small nanoparticles, smaller than 5–6 nm in diameter, are exclusively cleared from the body by renal filtration and urine excretion [47]. Slightly larger particles, 10–100 nm in diameter, that escape the renal filtration face the hepatobiliary system and tend to accumulate in the liver by passing through the vascular fenestration measuring 50–100 nm [48]. The splenic macrophages have a significant role in phagocytosis of nanoparticles larger than 200 nm and, lastly, particles in micrometer range become trapped in the capillaries of the lung and mainly accumulate in this organ [49-52].
Various geometries of nanoparticles affect pharmacokinetics and biodistribution of nanoparticles. For instance, cylindrical and discoidal nanoparticles have unique flow characteristics, the former prone to tumbling and margination dynamics favoring nanoparticles binding and adhesion to endothelium [53, 54] and the latter prone to long circulation time owing to the alignment of cylinders in the vessels with blood flow [55, 56]. Another important feature in designing nanoparticles with prolonged circulation time and selective organ accumulation is surface charge [57, 58]. Nanoparticles with neutral or negative surface charge have longer blood circulation time because of less nonspecific protein adsorption and opsonization [59, 60]. On one hand, positively charged nanoparticles accumulate non-specifically in most of the organs, whereas, on the other hand, negatively charged nanoparticles accumulate less in the liver and spleen [61]. Positively charged nanoparticles have been shown to preferentially be uptaken by tumor-associated angiogenic endothelial cells and facilitate tumor accumulation [45, 62, 63].
MECHANISM OF ORGAN DELIVERY
Passive delivery
Developing nanocarriers with prolonged circulation time revolutionized the field of nanomedicine about 30 years ago since nanocarriers would passively accumulate at a particulate site [64]. The passive accumulation solemnly depends on hemodynamics of blood and diffusion, and disrupted endothelia, the need for targeting ligands on the surface of nanoparticles. In the field of cancer nanomedicine, the majority of nanomedicine application relies on the passive targeting of the tumor vasculature [65, 66]. Owing to the leakiness of the tumor vasculature, particles of appropriate size may accumulate passively through EPR effect [26, 67]. Excessive branching of the vasculature in the tumor combined with enlarged gap between the endothelial cells, replacing the tight junctions, has displayed facilitated extravasation of nanocarriers to the tumor site [68, 69].
Several research groups have reported using nanocarriers for delivery of immune therapeutics [70■■, 71■■]. Passive delivery of tacrolimus to the lymphatics using PLGA nanoparticles with average size of 200 nm was reported by Shin et al. [72]. They showed that intravenously administered tacrolimus-loaded nanoparticles accumulate at the lymphatic sites by passing through the lymphatic capillary fenestrations, although no therapeutic effect was demonstrated [72]. In another study, PEGylated Poly(lactic acid) nanoparticle loaded with tacrolimus (100 nm diameter) were studied in a rat liver transplant model. The tacrolimus-loaded nanoparticles were delivered through gastric perfusion and lead to significant graft survival prolongation [73].
Liposomal cyclosporine nanoparticles were another type of nanoparticles investigated in liver transplantation, showing reduced off-target side-effects in a rat transplant model as nanocarriers trafficked at a higher rate to the liver as the main member of reticuloendothelial system [74].
Look et al. [75] have reported targeting dendritic cells ex vivo using nanogel-based nanoparticles and solid PLGA nanoparticles. Nanogels loaded with mycophenolic acid showed higher internalization efficacy by dendritic cells and greater immunosuppression. Immunosuppressive therapy of lupus-prone New Zealand Black/White F1 Hybrid mice with nanogels resulted in extended survival of the animals. Several research groups have investigates encapsulation of rapamycin in polymeric particles in vitro. They have reported sustained release of rapamycin over several days [poly(ethylene glycol)-block-poly(caprolactone) micelles] [76], resulting in superior inhibition of dendritic cells maturation [77, 78], and induction of regulatory T cells (Treg) induction by combinational delivery of interleukin-2, transforming growth factor-β, and rapamycin [79].
Targeted delivery
Active targeting is the other method that relies on selective interaction of ligands on the surface of particles with the receptor of interest at the target site [80, 81]. Therefore, the particles are internalized by cells through receptor-mediated endocytosis and deliver their payload at the site of interest [82, 83]. Surface conjugation modality widens the field of application of nanocarriers in medicine. Antibodies, peptides, proteins, small molecules, low molecular weight ligands, and polysaccharides are some examples of targeting moieties to conjugate to the surface of nanoparticles [82-86].
Targeted drug delivery in transplantation
Given the pathobiology of transplant rejection, two sites of lymphoid tissue and organ transplant are of interest for the delivery of immune therapeutics. There have been efforts for the direct delivery of molecules to the organs to suppress early inflammatory responses [87, 88■■]. However, the focus of this review is on the delivery of immune therapeutics to lymph nodes following systemic administration.
Lymph node delivery of immune therapeutics
The lymph node has been the focus of transplantation research for decades. Lymph nodes are well recognized sites where the alloantigens are introduced to the T cells by dendritic cells. The microarchitecture of lymph nodes is critical to their function [89]. The seminal discovery of Sir James Gowans showing the daily homing of lymphocytes to the lymph nodes has shaped our understanding of immune activation in the lymph node [90-92]. The ability of immune therapeutics to reach the lymph node creates an unprecedented opportunity to augment or diminish the immune activity for the disease of interest. One can envision development of more effective vaccines or enhanced tumor immunity by delivering the antigens or therapeutics of interest to further augment the immune response. The DLN is an ideal target for delivery of immune therapeutics in transplantation as the primary site of alloreactive T-cell activation [93, 94]. Thus far, most of such deliveries solely rely on the subcutaneous injection of materials to reach the DLN of interest.
Subcutaneous injection is an extensively studied route of administration of therapeutic carriers, specifically liposomes, for delivery to the lymph node [95]. Size of the liposomes and the injection site are the most important factors influencing lymph node targeting via subcutaneous injection. Phagocytosis by macrophages is the driving force behind the targeted delivery of liposomes to the regional lymph nodes [95-99]. Dane et al. [100] have encapsulated tacrolimus inside micelles with average diameter of 50 nm, using amphiphilic poly(ethylene glycol)-bl-poly(propylene sulfide) block copolymers. The efficacy of tacrolimus micelles was investigated in the allogenic C57BL/6 tail skin transplantation model and graft survival prolongation was only achieved when tacrolimus/rapamycin micelles combinational therapy was used.
Azzi et al. [101] designed polylactide–cyclosporine A nanocarriers and coupled them ex vivo to dendritic cells for targeted drug delivery to the lymph nodes. The dendritic cell–nanocarrier combination injected into the footpads of mice showed enhanced drug delivery and suppression of T-cell proliferation in the lymph nodes.
Active targeted delivery to the lymph node
On the contrary, delivery of molecules to the lymph nodes following systemic administration has been scarce, thus far, as it is a more daunting task compared with passive delivery. There is a need for pragmatic approaches that would allow delivery to the deeper chain of lymph nodes in the body. Visceral lymph nodes are primary sites of interest, for instance, not only for transplantation but also for cancer therapy. Lymphocytes interact with specialized vasculature present in lymph nodes referred to as high endothelial venules (HEVs). This vasculature was long recognized by their bubbly appearance as compared to the conventional venular cells that are flat [102]. L-selectin present on naïve T cells interacts with a series of sugar-coated molecules called peripheral node addressing (PNAd) molecules [103]. PNAd molecules are collectively recognized by a monoclonal antibody called MECA-79.
We have designed microparticles (MPs) which can follow the foot steps of lymphocytes. MPs containing tacrolimus were coated with MECA-79 that specifically homed to the DLN. Our study indicates that such delivery allows to achieve prolongation of heart allograft survival with much lower circulatory levels of tacrolimus [104■■]. There is a need for future studies to improve the trafficking of nanoparticles to lymph nodes. The efficacy of such delivery in various disease models needs to be tested. Furthermore, transplant organs suffering from chronic rejection develop tertiary lymphoid tissue (TLO) that share many features of lymph node including PNAd expressing HEVs. Pathogenic effector memory T cells residing in TLO play a key role in the pathogenesis of chronic rejection [105-112]. Interestingly, blocking TLO reduces chronic rejection [113]. Again, our HEV nanodelivery platform enables the delivery of immune therapeutics to TLO to target pathogenic T cells. Moreover, our delivery of immune therapeutics to transplant organs has the potential to inhibit both inflammatory responses and effector and memory T cells within the organ.
CONCLUSION
The exploitation of innovative strategies for the active targeted delivery of immune therapeutics to the DLN in transplantation is a crucial clinical necessity. Despite rapid expansion of nanomedicine in the field of cancer therapy, less is explored in immunotherapy and transplantation. Therefore, more studies are needed to identify targeting molecules and effective delivery platforms without posing systemic toxicity.
KEY POINTS.
Targeted delivery of immune therapeutics using nanoparticles has great potential for developing systems to achieve desired immune response.
Immune therapeutics–nanoparticle drug delivery platforms can enhance the therapeutic index of immune therapeutics along with lowered systemic toxicity.
Immune therapeutics–nanoparticles platform carries considerable potential to deliver a wide variety of immune therapeutic molecules.
Targeted delivery of immune therapeutics to draining lymph node (DLN) and the transplanted organ has the potential to suppress immune response as well as inhibit the inflammatory response.
Acknowledgments
Financial support and sponsorship
This work was supported by the National Institute of Allergy and Infectious Diseases of the National Institutes of Health under Award Number RO1AI126596 and K24AI116925 (R.A.).
Footnotes
Conflicts of interest
The authors have no conflicts of interest.
REFERENCES AND RECOMMENDED READING
Papers of particular interest, published within the annual period of review, have been highlighted as:
■ of special interest
■■ of outstanding interest
- 1.Ojo AO, Hanson JA, Wolfe RA, et al. Long-term survival in renal transplant recipients with graft function. Kidney Int 2000; 57:307–313. [DOI] [PubMed] [Google Scholar]
- 2.Vincenti F Immunosuppression minimization: current and future trends in transplant immunosuppression. J Am Soc Nephrol 2003; 14:1940–1948. [DOI] [PubMed] [Google Scholar]
- 3.Naesens M, Kuypers DR, Sarwal M. Calcineurin inhibitor nephrotoxicity. Clin J Am Soc Nephrol 2009; 4:481–508. [DOI] [PubMed] [Google Scholar]
- 4.Vincenti F, Larsen C, Durrbach A, et al. Costimulation blockade with belatacept in renal transplantation. N Engl J Med 2005; 353:770–781. [DOI] [PubMed] [Google Scholar]
- 5.Meier-Kriesche HU, Baliga R, Kaplan B. Decreased renal function is a strong risk factor for cardiovascular death after renal transplantation. Transplantation 2003; 75:1291–1295. [DOI] [PubMed] [Google Scholar]
- 6.Halloran PF. Immunosuppressive drugs for kidney transplantation. N Engl J Med 2004; 351:2715–2729. [DOI] [PubMed] [Google Scholar]
- 7.Naesens M, Lerut E, Sarwal M, et al. Balancing efficacy and toxicity of kidney transplant immunosuppression. Transplant Proc 2009; 41:3393–3395. [DOI] [PubMed] [Google Scholar]
- 8.Euvrard S, Morelon E, Rostaing L, et al. Sirolimus and secondary skin-cancer prevention in kidney transplantation. N Engl J Med 2012; 367:329–339. [DOI] [PubMed] [Google Scholar]
- 9.Vajdic CM, van Leeuwen MT. Cancer incidence and risk factors after solid organ transplantation. Int J Cancer 2009; 125:1747–1754. [DOI] [PubMed] [Google Scholar]
- 10.Mitchell RN. Graft vascular disease: immune response meets the vessel wall. Annu Rev Pathol 2009; 4:19–47. [DOI] [PubMed] [Google Scholar]
- 11.Prakash J, Ghosh B, Singh S, et al. Causes of death in renal transplant recipients with functioning allograft. Indian J Nephrol 2012; 22:264–268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Svensson M, Jardine A, Fellstrom B, Holdaas H. Prevention of cardiovascular disease after renal transplantation. Curr Opin Organ Transplant 2012; 17:393–400. [DOI] [PubMed] [Google Scholar]
- 13.Pilmore HL, Skeans MA, Snyder JJ, et al. Cardiovascular disease medications after renal transplantation: results from the Patient Outcomes in Renal Transplantation study. Transplantation 2011; 91:542–551. [DOI] [PubMed] [Google Scholar]
- 14.Engels EA, Pfeiffer RM, Fraumeni JF Jr, et al. Spectrum of cancer risk among US solid organ transplant recipients. JAMA 2011; 306:1891–1901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Flechner SM, Kobashigawa J, Klintmalm G. Calcineurin inhibitor-sparing regimens in solid organ transplantation: focus on improving renal function and nephrotoxicity. Clin Transplant 2008; 22:1–15. [DOI] [PubMed] [Google Scholar]
- 16.Haller M, Oberbauer R. Calcineurin inhibitor minimization, withdrawal and avoidance protocols after kidney transplantation. Transpl Int 2009; 22:69–77. [DOI] [PubMed] [Google Scholar]
- 17.Srinivas TR, Meier-Kriesche HU. Minimizing immunosuppression, an alternative approach to reducing side effects: objectives and interim result. Clin J Am Soc Nephrol 2008; 3(Suppl 2):S101–S116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Perez-Medina C, Hak S, Reiner T, et al. Integrating nanomedicine and imaging. Philos Trans A Math Phys Eng Sci 2017; 375:pii: 20170110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Jurj A, Braicu C, Pop LA, et al. The new era of nanotechnology, an alternative to change cancer treatment. Drug Des Devel Ther 2017; 11:2871–2890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Barkalina N, Charalambous C, Jones C, Coward K. Nanotechnology in reproductive medicine: emerging applications of nanomaterials. Nanomedicine 2014; 10:921–938. [DOI] [PubMed] [Google Scholar]
- 21.Ramos AP, Cruz MAE, Tovani CB, Ciancaglini P. Biomedical applications of nanotechnology. Biophys Rev 2017; 9:79–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Farokhzad OC, Langer R. Impact of nanotechnology on drug delivery. ACS Nano 2009; 3:16–20. [DOI] [PubMed] [Google Scholar]
- 23.Shi J, Xiao Z, Kamaly N, Farokhzad OC. Self-assembled targeted nanoparticles: evolution of technologies and bench to bedside translation. Acc Chem Res 2011; 44:1123–1134. [DOI] [PubMed] [Google Scholar]
- 24.Wang AZ, Gu F, Zhang L, et al. Biofunctionalized targeted nanoparticles for therapeutic applications. Expert Opin Biol Ther 2008; 8:1063–1070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Park BH, Hwang T, Liu TC, et al. Use of a targeted oncolytic poxvirus, JX-594, in patients with refractory primary or metastatic liver cancer: a phase I trial. Lancet Oncol 2008; 9:533–542. [DOI] [PubMed] [Google Scholar]
- 26.Maeda H Tumor-selective delivery of macromolecular drugs via the EPR effect: background and future prospects. Bioconjug Chem 2010; 21:797–802. [DOI] [PubMed] [Google Scholar]
- 27.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 Res 1986; 46(12 Pt 1):6387–6392. [PubMed] [Google Scholar]
- 28.Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases. Nature 2000; 407:249–257. [DOI] [PubMed] [Google Scholar]
- 29.Kamaly N, Xiao Z, Valencia PM, et al. Targeted polymeric therapeutic nanoparticles: design, development and clinical translation. Chem Soc Rev 2012; 41:2971–3010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Torchilin VP. Recent advances with liposomes as pharmaceutical carriers. Nat Rev Drug Discov 2005; 4:145–160. [DOI] [PubMed] [Google Scholar]
- 31.Mitragotri S, Burke PA, Langer R. Overcoming the challenges in administering biopharmaceuticals: formulation and delivery strategies. Nat Rev Drug Discov 2014; 13:655–672. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Chan JM, Valencia PM, Zhang L, et al. Polymeric nanoparticles for drug delivery. Methods Mol Biol 2010; 624:163–175. [DOI] [PubMed] [Google Scholar]
- 33.Yameen B, Choi WI, Vilos C, et al. Insight into nanoparticle cellular uptake and intracellular targeting. J Control Release 2014; 190:485–499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Wang Z, Malik AB. Nanoparticles squeezing across the blood–endothelial barrier via caveolae. Ther Deliv 2013; 4:131–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Georgieva JV, Kalicharan D, Couraud PO, et al. Surface characteristics of nanoparticles determine their intracellular fate in and processing by human blood–brain barrier endothelial cells in vitro. Mol Ther 2011; 19:318–325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Ferrari M Frontiers in cancer nanomedicine: directing mass transport through biological barriers. Trends Biotechnol 2010; 28:181–188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Kamaly N, Yameen B, Wu J, Farokhzad OC. Degradable controlled-release polymers and polymeric nanoparticles: mechanisms of controlling drug release. Chem Rev 2016; 116:2602–2663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Rivera E Current status of liposomal anthracycline therapy in metastatic breast cancer. Clin Breast Cancer 2003; 4(Suppl 2):S76–S83. [DOI] [PubMed] [Google Scholar]
- 39.Hofheinz RD, Gnad-Vogt SU, Beyer U, Hochhaus A. Liposomal encapsulated anticancer drugs. Anticancer Drugs 2005; 16:691–707. [DOI] [PubMed] [Google Scholar]
- 40.Wu J, Liu Q, Lee RJ. A folate receptor-targeted liposomal formulation for paclitaxel. Int J Pharm 2006; 316:148–153. [DOI] [PubMed] [Google Scholar]
- 41.Cho SW, Goldberg M, Son SM, et al. Lipid-like nanoparticles for small interfering RNA delivery to endothelial cells. Adv Funct Mater 2009; 19:3112–3118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Danhier F, Ansorena E, Silva JM, et al. PLGA-based nanoparticles: an overview of biomedical applications. J Control Release 2012; 161:505–522. [DOI] [PubMed] [Google Scholar]
- 43.Biswas S, Kumari P, Lakhani PM, Ghosh B. Recent advances in polymeric micelles for anticancer drug delivery. Eur J Pharm Sci 2015; 83:184–202. [DOI] [PubMed] [Google Scholar]
- 44.Ernsting MJ, Murakami M, Roy A, Li SD. Factors controlling the pharmacokinetics, biodistribution and intratumoral penetration of nanoparticles. J Control Release 2013; 172:782–794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Li Y, Pei Y, Zhang X, et al. PEGylated PLGA nanoparticles as protein carriers: synthesis, preparation and biodistribution in rats. J Control Release 2001; 71:203–211. [DOI] [PubMed] [Google Scholar]
- 46.He C, Hu Y, Yin L, et al. Effects of particle size and surface charge on cellular uptake and biodistribution of polymeric nanoparticles. Biomaterials 2010; 31:3657–3666. [DOI] [PubMed] [Google Scholar]
- 47.Choi HS, Liu W, Misra P, et al. Renal clearance of quantum dots. Nat Biotechnol 2007; 25:1165–1170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Braet F, Wisse E, Bomans P, et al. Contribution of high-resolution correlative imaging techniques in the study of the liver sieve in three-dimensions. Microsc Res Tech 2007; 70:230–242. [DOI] [PubMed] [Google Scholar]
- 49.Longmire M, Choyke PL, Kobayashi H. Clearance properties of nano-sized particles and molecules as imaging agents: considerations and caveats. Nanomedicine (Lond) 2008; 3:703–717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Owens DE 3rd, Peppas NA. Opsonization, biodistribution, and pharmacokinetics of polymeric nanoparticles. Int J Pharm 2006; 307:93–102. [DOI] [PubMed] [Google Scholar]
- 51.Otsuka H, Nagasaki Y, Kataoka K. PEGylated nanoparticles for biological and pharmaceutical applications. Adv Drug Delivery Rev 2003; 55:403–419. [DOI] [PubMed] [Google Scholar]
- 52.Moghimi SM, Szebeni J. Stealth liposomes and long circulating nanoparticles: critical issues in pharmacokinetics, opsonization and protein-binding properties. Prog Lipid Res 2003; 42:463–478. [DOI] [PubMed] [Google Scholar]
- 53.Gentile F, Chiappini C, Fine D, et al. The effect of shape on the margination dynamics of nonneutrally buoyant particles in two-dimensional shear flows. J Biomech 2008; 41:2312–2318. [DOI] [PubMed] [Google Scholar]
- 54.Black KC, Wang Y, Luehmann HP, et al. Radioactive 198Au-doped nanostructures with different shapes for in vivo analyses of their biodistribution, tumor uptake, and intratumoral distribution. ACS Nano 2014; 8:4385–4394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Geng Y, Dalhaimer P, Cai S, et al. Shape effects of filaments versus spherical particles in flow and drug delivery. Nat Nanotechnol 2007; 2:249–255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Liu X, Wu F, Tian Y, et al. Size dependent cellular uptake of rod-like bionanoparticles with different aspect ratios. Sci Rep 2016; 6:24567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Mout R, Moyano DF, Rana S, Rotello VM. Surface functionalization of nanoparticles for nanomedicine. Chem Soc Rev 2012; 41:2539–12539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Kobayashi K, Wei J, Iida R, et al. Surface engineering of nanoparticles for therapeutic applications. Polymer J 2014; 46:460–468. [Google Scholar]
- 59.Alexis F, Pridgen E, Molnar LK, Farokhzad OC. Factors affecting the clearance and biodistribution of polymeric nanoparticles. Mol Pharm 2008; 5:505–515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Dobrovolskaia MA, Aggarwal P, Hall JB, McNeil SE. Preclinical studies to understand nanoparticle interaction with the immune system and its potential effects on nanoparticle biodistribution. Mol Pharm 2008; 5:487–495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Yamamoto Y, Nagasaki Y, Kato Y, et al. Long-circulating poly(ethylene glycol)-poly(D,L-lactide) block copolymer micelles with modulated surface charge. J Control Release 2001; 77:27–38. [DOI] [PubMed] [Google Scholar]
- 62.Thurston G, McLean JW, Rizen M, et al. Cationic liposomes target angiogenic endothelial cells in tumors and chronic inflammation in mice. J Clin Invest 1998; 101:1401–1413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Gref R, Lück M, Quellec P, et al. ‘Stealth’ corona-core nanoparticles surface modified by polyethylene glycol (PEG): influences of the corona (PEG chain length and surface density) and of the core composition on phagocytic uptake and plasma protein adsorption. Colloids Surf B Biointerfaces 2000; 18:301–313. [DOI] [PubMed] [Google Scholar]
- 64.Danhier F, Feron O, Preat V. To exploit the tumor microenvironment: passive and active tumor targeting of nanocarriers for anticancer drug delivery. J Control Release 2010; 148:135–146. [DOI] [PubMed] [Google Scholar]
- 65.Bertrand N, Wu J, Xu X, et al. Cancer nanotechnology: the impact of passive and active targeting in the era of modern cancer biology. Adv Drug Deliv Rev 2014; 66:2–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Noguchi Y, Wu J, Duncan R, et al. Early phase tumor accumulation of macromolecules: a great difference in clearance rate between tumor and normal tissues. Jpn J Cancer Res 1998; 89:307–314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Azzopardi EA, Ferguson EL, Thomas DW. The enhanced permeability retention effect: a new paradigm for drug targeting in infection. J Antimicrob Chemother 2013; 68:257–274. [DOI] [PubMed] [Google Scholar]
- 68.Jain RK. The next frontier of molecular medicine: delivery of therapeutics. Nat Med 1998; 4:655–657. [DOI] [PubMed] [Google Scholar]
- 69.Jain RK, Stylianopoulos T. Delivering nanomedicine to solid tumors. Nat Rev Clin Oncol 2010; 7:653–664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.■■.Rincon-Restrepo M, Mayer A, Hauert S, et al. Vaccine nanocarriers: coupling intracellular pathways and cellular biodistribution to control CD4 vs CD8 T cell responses. Biomaterials 2017; 132:48–58.This study reports application of nanoparticulates in vaccine design and its effect on adaptive immune response.
- 71.■■.Baekkeskov S, Hubbell JA, Phelps EA. Bioengineering strategies for inducing tolerance in autoimmune diabetes. Adv Drug Deliv Rev 2017; 114:256–265.This review article has summarized the recent findings in bioengineering toward type 1 diabetes treatment.
- 72.Shin SB, Cho HY, Kim DD, et al. Preparation and evaluation of tacrolimus-loaded nanoparticles for lymphatic delivery. Eur J Pharm Biopharm 2010; 74:164–171. [DOI] [PubMed] [Google Scholar]
- 73.Xu W, Ling P, Zhang T. Toward immunosuppressive effects on liver transplantation in rat model: tacrolimus loaded poly(ethylene glycol)-poly(D,L-lactide) nanoparticle with longer survival time. Int J Pharm 2014; 460:173–180. [DOI] [PubMed] [Google Scholar]
- 74.Freise CE, Liu T, Hong K, et al. The increased efficacy and decreased nephrotoxicity of a cyclosporine liposome. Transplantation 1994; 57:928–932. [DOI] [PubMed] [Google Scholar]
- 75.Look M, Saltzman WM, Craft J, Fahmy TM. The nanomaterial-dependent modulation of dendritic cells and its potential influence on therapeutic immunosuppression in lupus. Biomaterials 2014; 35:1089–1095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Forrest ML, Won CY, Malick AW, Kwon GS. In vitro release of the mTOR inhibitor rapamycin from poly(ethylene glycol)-b-poly(e-caprolactone) micelles. J Control Release 2006; 110:370–377. [DOI] [PubMed] [Google Scholar]
- 77.Lewis JS, Roche C, Zhang Y, et al. Combinatorial delivery of immunosuppressive factors to dendritic cells using dual-sized microspheres. J Mater Chem B Mater Biol Med 2014; 2:2562–2574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Haddadi A, Elamanchili P, Lavasanifar A, et al. Delivery of rapamycin by PLGA nanoparticles enhances its suppressive activity on dendritic cells. J Biomed Mater Res A 2008; 84A:885–898. [DOI] [PubMed] [Google Scholar]
- 79.Jhunjhunwala S, Balmert SC, Raimondi G, et al. Controlled release formulations of IL-2, TGF-β1 and rapamycin for the induction of regulatory T cells. J Control Release 2012; 159:78–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Yu MK, Park J, Jon S. Targeting strategies for multifunctional nanoparticles in cancer imaging and therapy. Theranostics 2012; 2:3–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Foster C, Watson A, Kaplinsky J, Kamaly N. Improved targeting of cancers with nanotherapeutics. Methods Mol Biol 2017; 1530:13–37. [DOI] [PubMed] [Google Scholar]
- 82.Yu B, Tai HC, Xue W, et al. Receptor-targeted nanocarriers for therapeutic delivery to cancer. Mol Membr Biol 2010; 27:286–298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Kolhatkar R, Lote A, Khambati H. Active tumor targeting of nanomaterials using folic acid, transferrin and integrin receptors. Curr Drug Discov Technol 2011; 8:197–206. [DOI] [PubMed] [Google Scholar]
- 84.des Rieux A, Pourcelle V, Cani PD, et al. Targeted nanoparticles with novel nonpeptidic ligands for oral delivery. Adv Drug Deliv Rev 2013; 65:833–844. [DOI] [PubMed] [Google Scholar]
- 85.Wang J, Tian S, Petros RA, et al. The complex role of multivalency in nanoparticles targeting the transferrin receptor for cancer therapies. J Am Chem Soc 2010; 132:11306–11313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Farokhzad OC, Cheng J, Teply BA, et al. Targeted nanoparticle-aptamer bioconjugates for cancer chemotherapy in vivo. Proc Natl Acad Sci USA 2006; 103:6315–6320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Solhjou Z, Athar H, Xu Q, Abdi R. Emerging therapies targeting intra-organ inflammation in transplantation. Am J Transplant 2015; 15:305–311. [DOI] [PubMed] [Google Scholar]
- 88.■■.Solhjou Z, Uehara M, Bahmani B, et al. Novel application of localized nanodelivery of anti-interleukin-6 protects organ transplant from ischemia-reperfusion injuries. Am J Transplant 2017; 17:2326–2337.This study reports direct intraorgan drug delivery and the effect on the immune response in organ transplantation.
- 89.Brinkman CC, Burrell BE, Iwami D, et al. Anatomy of tolerance. Curr Opin Organ Transplant 2013; 18:393–401. [DOI] [PubMed] [Google Scholar]
- 90.Butcher EC, Picker LJ. Lymphocyte homing and homeostasis. Science 1996; 272:60–66. [DOI] [PubMed] [Google Scholar]
- 91.Jalkanen S, Reichert RA, Gallatin WM, et al. Homing receptors and the control of lymphocyte migration. Immunol Rev 1986; 91:39–60. [DOI] [PubMed] [Google Scholar]
- 92.Marchesi VT, Gowans JL. The migration of lymphocytes through the endothelium of venules in lymph nodes: an electron microscope study. Proc R Soc Lond B Biol Sci 1964; 159:283–290. [DOI] [PubMed] [Google Scholar]
- 93.Lakkis FG, Sayegh MH. Memory T cells: a hurdle to immunologic tolerance. J Am Soc Nephrol 2003; 14:2402–2410. [DOI] [PubMed] [Google Scholar]
- 94.Park CO, Kupper TS. The emerging role of resident memory T cells in protective immunity and inflammatory disease. Nat Med 2015; 21:688–697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Kim H, Kim Y, Lee J. Liposomal formulations for enhanced lymphatic drug delivery. Asian J Pharmaceutic Sci 2013; 8:96–103. [Google Scholar]
- 96.Oussoren C, Velinova M, Scherphof G, et al. Lymphatic uptake and biodistribution of liposomes after subcutaneous injection. IV. Fate of liposomes in regional lymph nodes. Biochim Biophys Acta 1998; 1370:259–272. [DOI] [PubMed] [Google Scholar]
- 97.Oussoren C, Zuidema J, Crommelin DJ, Storm G. Lymphatic uptake and biodistribution of liposomes after subcutaneous injection. II. Influence of liposomal size, lipid compostion and lipid dose. Biochim Biophys Acta 1997; 1328:261–272. [DOI] [PubMed] [Google Scholar]
- 98.Oussoren C, Storm G. Lymphatic uptake and biodistribution of liposomes after subcutaneous injection: III. Influence of surface modification with poly(ethyleneglycol). Pharm Res 1997; 14:1479–1484. [DOI] [PubMed] [Google Scholar]
- 99.Oussoren C, Storm G. Liposomes to target the lymphatics by subcutaneous administration. Adv Drug Deliv Rev 2001; 50:143–156. [DOI] [PubMed] [Google Scholar]
- 100.Dane KY, Nembrini C, Tomei AA, et al. Nano-sized drug-loaded micelles deliver payload to lymph node immune cells and prolong allograft survival. J Control Release 2011; 156:154–160. [DOI] [PubMed] [Google Scholar]
- 101.Azzi J, Tang L, Moore R, et al. Polylactide-cyclosporin A nanoparticles for targeted immunosuppression. FASEB J 2010; 24:3927–3938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Girard JP, Springer TA. High endothelial venules (HEVs): specialized endothelium for lymphocyte migration. Immunol Today 1995; 16:449–457. [DOI] [PubMed] [Google Scholar]
- 103.von Andrian UH, Mackay CR. T-cell function and migration. Two sides of the same coin. N Engl J Med 2000; 343:1020–1034. [DOI] [PubMed] [Google Scholar]
- 104. ■■.Azzi J, Yin Q, Uehara M, et al. Targeted delivery of immunomodulators to lymph nodes. Cell Rep 2016; 15:1202–1213.This study reports the first targeted immune therapeutics delivery to the lymph node.
- 105.Neyt K, Perros F, GeurtsvanKessel CH, et al. Tertiary lymphoid organs in infection and autoimmunity. Trends Immunol 2012; 33:297–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Michie SA, Streeter PR, Bolt PA, et al. The human peripheral lymph node vascular addressin. An inducible endothelial antigen involved in lymphocyte homing. Am J Pathol 1993; 143:1688–1698. [PMC free article] [PubMed] [Google Scholar]
- 107.Xu B, Wagner N, Pham LN, et al. Lymphocyte homing to bronchus-associated lymphoid tissue (BALT) is mediated by L-selectin/PNAd, alpha4beta1 integrin/VCAM-1, and LFA-1 adhesion pathways. J Exp Med 2003; 197:1255–1267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Baddoura FK, Nasr IW, Wrobel B, et al. Lymphoid neogenesis in murine cardiac allografts undergoing chronic rejection. Am J Transplant 2005; 5:510–516. [DOI] [PubMed] [Google Scholar]
- 109.Yin N, Zhang N, Xu J, et al. Targeting lymphangiogenesis after islet transplantation prolongs islet allograft survival. Transplantation 2011; 92:25–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Nasr IW, Reel M, Oberbarnscheidt MH, et al. Tertiary lymphoid tissues generate effector and memory T cells that lead to allograft rejection. Am J Transplant 2007; 7:1071–1079. [DOI] [PubMed] [Google Scholar]
- 111.Krupnick AS, Lin X, Li W, et al. Orthotopic mouse lung transplantation as experimental methodology to study transplant and tumor biology. Nat Protoc 2009; 4:86–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Sato M, Hirayama S, Hwang DM, et al. The role of intrapulmonary de novo lymphoid tissue in obliterative bronchiolitis after lung transplantation. J Immunol 2009; 182:7307–7316. [DOI] [PubMed] [Google Scholar]
- 113.Motallebzadeh R, Rehakova S, Conlon TM, et al. Blocking lymphotoxin signaling abrogates the development of ectopic lymphoid tissue within cardiac allografts and inhibits effector antibody responses. FASEB J 2012; 26:51–62. [DOI] [PubMed] [Google Scholar]
