key: cord-017583-72mbsib7 authors: Devarajan, Padma V.; Dawre, Shilpa M.; Dutta, Rinku title: Infectious Diseases: Need for Targeted Drug Delivery date: 2014-09-01 journal: Targeted Drug Delivery : Concepts and Design DOI: 10.1007/978-3-319-11355-5_3 sha: doc_id: 17583 cord_uid: 72mbsib7 Infectious diseases are a leading cause of death worldwide, with the constant fear of global epidemics. It is indeed an irony that the reticuloendothelial system (RES), the body’s major defence system, is the primary site for intracellular infections which are more difficult to treat. Pro-inflammatory M1 macrophages play an important role in defence. However, ingenious pathogen survival mechanisms including phagolysosome destruction enable their persistence. Microbial biofilms present additional challenges. Low intracellular drug concentrations, drug efflux by efflux pumps and/or enzymatic degradation, emergence of multi-drug resistance (MDR), are serious limitations of conventional therapy. Targeted delivery using nanocarriers, and passive and active targeting strategies could provide quantum increase in intracellular drug concentration. Receptor mediated endocytosis using appropriate ligands is a viable approach. Liposomes and polymeric/lipidic nanoparticles, dendrimers micelles and micro/nanoemulsions could all be relied upon. Specialised targeting approaches are demonstrated for important diseases like tuberculosis, HIV and Malaria. Application of targeted delivery in the treatment of veterinary infections is exemplified and future possibilities indicated. The chapter thus provides an overview on important aspects of infectious diseases and the challenges therein, while stressing on the promise of targeted drug delivery in augmenting therapy of infectious diseases. of the microorganism. Intracellular infections result when the organisms cleverly evade destruction following phagocytosis. The intracellular location of these microorganisms protects them from the host defence mechanisms, such as antibodies or complement, and from the action of drugs that are unable to penetrate the cell effi ciently. Hence, while adequate drug concentrations are readily achieved at extracellular infection sites to enable effi cient therapy, intracellular infections are more diffi cult to treat. Some common intracellular and extracellular infectious diseases and their causative organisms are listed in Table 3 .1 . The RES also known as the mononuclear phagocytic system (MPS)/macrophage system is the primary defence mechanism of the human body and hence the site of intracellular infections. The macrophages constitute the major defence cells of the RES. Derived from the bone marrow the RES also contributes to both non-specifi c and specifi c immunity. Recognition by the RES is facilitated by opsonins, with the step of opsonisation being a precursor to phagocytosis. Opsonisation is the process by which bacteria are altered by opsonins so as to become more readily and effi ciently engulfed by phagocytosis. Opsonisation is mediated by the complement system: C3b, C4b, and iC3b, antibodies IgG and IgM and mannose-binding lectin. Mannose binding lectin initiates the formation of C3b. Opsonisation of particles enables recognition by the Fc receptors, complement receptors or specifi c receptors for phagocytosis. Opsonins are generally proteins which can bind to pattern-recognition receptors (PRRs) or other specifi c receptors expressed on the surface of macrophages. Pentraxins [C-reactive protein and serum amyloid P] [ 1 ] , mindin, collectins [ 2 ] and fi colins [ 3 ] are such opsonins. The function of pattern-recognition receptors (PRRs) is to recognise and enhance phagocytosis of pathogen-associated molecular patterns (PAMPs), specifi c patterns present on microbial pathogens like lipopolysaccharide (LPS) in Gram-negative bacteria, lipotechoic acid (LTA) in Gram-positive bacteria and mannans in yeast. Toll-like receptors (TLRs) are PRRs essential for recognition of microbial components such as TLR4 (LPS) [ 4 -6 ] , TLR3 [double-stranded RNA] [ 7 ] , TLR6 [mycoplasmal macrophage-activating lipopeptide-2 kDa] [ 8 ] , TLR9 [CpG bacterial DNA] [ 9 ] , TLR5 [bacterial fl agellin] [ 10 ] , and TLR2 [peptidoglycan] . However, the exact mechanisms of TLR recognition of microbial components remain unclear. Opsonisation facilitates adherence of pathogens to macrophages, and is facilitated by integrins. Adherence induces membrane protrusions, called pseudopodia, to extend around the attached material. Following fusion with the macrophage, the pseudopodia forms a phagosome that encloses the pathogen within a membrane, which then enters the endocytic process. Phagosomes coalesce with intracellular organelles to mature into phagolysosomes, which have an acidic environment with many digestive proteins which fi nally degrades the internalised material. Phagocytised material is eliminated by exocytosis. The process of phagocytosis is mediated by several proteins such as actin, dynamin and cortactin. While actin is connected to the lipidic membrane and responsible for invagination of the membrane to form the endosome, cortactin is an actin-binding protein which stimulates its polymerisation. Dynamin hydrolyses guanidine triphosphate and uses the resulting energy for the contraction of actin and formation of endosome. Particulates that cannot be digested remain sequestered in residual bodies within the cell. Other cells such as fi broblast, endothelial and epithelial cells also exhibit phagocytic activity and can engulf microbes like Shigella, Listeria and Yersinia [ 11 ] . Such phagocytosis is mediated by laminin and fi bronectin receptors/heparan sulfate present on the membrane surface [ 11 ] . However, the major cells responsible for phagocytosis are macrophages. Macrophages (Greek: makros means large and phagein means eat) are cells formed by the differentiation of monocytes in tissues. Macrophages play an important role in both innate and adaptive immunity in vertebrates. These specialised phagocytic cells engulf and destroy infectious microbes, foreign particles and cancer cells [ 12 ] . The macrophages also regulate lymphocyte, granulocyte populations and important tumor growth modulators [ 13 ] . Macrophages act by both oxygen-dependent killing and oxygen independent killing mechanisms. The mediators for oxygen-dependent killing are reactive oxygen intermediates (ROIs) (superoxide anion, hydroxyl radicals, hydrogen peroxide and hypochlorite anion), reactive nitrogen intermediates (RNIs) (nitric oxide, nitrogen dioxide and nitrous acid) and monochloramine, while the mediators for oxygen independent killing are defensins, tumor necrosis factor (macrophage only), lysozyme and hydrolytic enzymes. Floating macrophages predominate in the vascular system, while tissue macrophages are localised in specifi c tissues. Based on the tissue of residence they have specifi c nomenclature ( Fig. 3.1 ). Macrophages can be classifi ed mainly into two groups: (1) pro-infl ammatory or classically activated macrophages (M1) and (2) anti-infl ammatory or alternatively activated macrophages (M2). M1 macrophages are immune effector cells that aggressively work against microbes and cause their destruction much more readily. M1 is mainly associated with gastrointestinal infections (e.g. typhoid fever and Helicobacter pylori gastritis) and active tuberculosis. M1 macrophages are stimulated by interferon (IFN)-g or lipopolysaccharide (LPS) to release nitric oxide (NO), important for killing intracellular pathogens. Activated macrophages are characterised by expression of major histocompatibility molecule like MHC class II and CD86 and their ability to secrete proinfl ammatory cytokines such as tumor necrosis factor (TNF)-a, IL-1b, IL-12, IL-18 and the chemokines CCL15, CCL20, CXCL8-11 and CXCL13 [ 14 ] . Activated M1 macrophages facilitate killing of microorganisms by endocytosis, synthesising reactive oxygen intermediates (ROI), limiting the uptake of nutrients and iron essential for the growth of bacteria and replication of viruses, or production of nitric oxide facilitated by IFN-g-inducible NO synthase (iNOS). M2 macrophages are important for killing extracellular parasites, wound healing, tissue repair, and to turn-off immune system activation. M2 macrophages are activated by interleukin (IL)-4 or IL-13 (M2a) to produce IL-10, transforming growth factor (TGF)-b and arginase-1 (Arg1), to enable this function [ 14 ] . M2 macrophages are mostly observed in lepromatous leprosy, Whipple's disease and localised infections (keratitis, chronic rhinosinusitis). A number of infectious organisms which manage to overcome the RES defence develop unique adaptive mechanisms which enable them to survive within the cell for prolonged periods of time. Eradication of such intracellular organisms poses immense challenges. Many pathogens have an innate ability to develop adaptive mechanisms under stress conditions to fi ght for their survival. Such adaptive mechanisms or protective strategies, enables them to exhibit greater defence to the host and there by prolong survival. The different adaptive mechanisms employed by pathogens are discussed below. Strategies adopted by microorganisms to inhibit phagolysosome formation include interference with the transformation of primary endosomes into late endosome, fusion with lysosomes and or phagosome acidifi cation. This delays the fusion of endosomes with lysosomes [ 15 ] or blocks the same [ 16 ] . The strategies to inhibit phagolysosome formation and the pathogens which exhibit the same [ 17 ] are summarised in Table 3 .2 . Pathogens which exhibit this adaptation survive and multiply in vesicles formed by fusion of endosomes with cell organelles other than the lysosome, such as the rough endoplasmic reticulum, ribosome or mitochondria [ 29 ] and thus avoid phagolysosome formation. They thereby bypass destruction due to the enzymatic activity in the lysosome [ 30 ] . Escape from endocytosis is a crucial step for intramacrophagic survival. Pathogens from this category contain lytic enzymes which enable them to break the endosomes membrane and disrupt membrane of the vacuole [ 31 ] , and hence evade degradation in the phagolysosome, and enter the cytosol rich in nutrients [ 32 ] . Specifi c enzymes are produced by the microorganisms for instance, L. monocytogenes Pujol et al. [ 17 ] Disturbs the formation of lipid rafts by producing beta-1,2 glucans Brucella spp. Brucellosis Roy [ 27 ] Alteration of host cell signaling by dephosphorylation of signal regulated kinase Leishmania spp. Leishmaniasis Ghosh et al. [ 28 ] produces listeriolysin O (LLO) [ 33 ] and haemolysin C [ 34 ] while phospholipases are produced by the Rickettsia spp. [ 35 ] . The microbes in this category exhibit virulence factors which allow them to survive in lytic enzymes, acidic conditions and oxidants, the harsh conditions in the phagolysosome environment. Intramacrophagic resistance employing multiple virulence factors enables alternative pathways for survival and multiplication [ 36 ] . Pathogens are internalised into macrophages by alternate routes. They traverse inside the cell by receptor mediated pathways like clathrin [ 37 ] and lipid rafts [ 38 ] . Formation of vesicles with new properties after fusion between the pathogen and membrane of the cell, like the parasitophorous vacuole formed by Toxoplasma gondii [ 38 ] also provides protection. In certain infections successful fusion of microorganisms with the macrophage is followed by secretion of antiapoptotic molecules (e.g. Bcl2). This results in impairment of apoptosis of the infected cells. In addition to the adaptive mechanisms certain microbes employ highly specifi c strategies for persistence inside the cell. Such strategies are discussed with reference to some important diseases. The adaptive mechanisms of Mycobacterium tuberculosis to survive inside the macrophages are prevention of fusion of the phagosome with lysosomes by producing tryptophan-aspartate-containing coat protein (TACO). Transformation of primary endosomes into phagolysosomes is prevented by a number of actions that occur simultaneously. These include reduced levels of proton ATPase inside the endosomes [ 18 ] and scavenger receptors [ 59 , 60 ] have also been implicated in mycobacterial uptake. Uptake of mycobacteria by the complement receptor pathway protects it from the aggressive lysosomal compartment ensuring relatively hospitable conditions. Salmonella specifi cally forms a glycolipid capsule or biofi lm. Biofi lm formation in salmonella is related to the multicellular and aggregative response of rdar [ 61 ] , rugose [ 62 ] , or lacy [ 63 ] . This multicellular behavior is a property of salmonellae [ 64 ] and is responsible for elaboration of thin fi mbriae like Tafi , curli [ 65 ] , cellulose [ 66 ] , and other uncharacterised extracellular polysaccharides. Together, these components form the extracellular matrix that confers resistance to acid and bleach and facilitates environmental persistence [ 62 , 64 , 67 -70 ] . Pathogens which cause fungal infections adapt various mechanisms to increase their pathogenesis and survive inside macrophages. C. albicans contains superoxide dismutases (SOD) and catalase enzymes which are able to convert O 2into molecular oxygen and hydrogen peroxide, thereby decreasing the scavenging and toxic effects of O 2and H 2 O 2 levels by certain reactions [ 71 ] . Further, C. neoformans evade phagocytic uptake by phenotypic switching. This mechanism is observed in yeast cells that express glucuronoxylomannan mucoid capsule that resist phagocytic uptake and cause high lethality in mice [ 72 ] . In case of Aspergillus conidia infection collectins, pentraxin proteins are essential for opsonisation, but their defi ciency is responsible for high susceptibility to infection in immunocompetent mice. Furthermore, several enzymes such as elastases and proteases released by the fungus enable conidia to escape from phagocytic uptake by alveolar macrophages. In HIV-1-infected macrophages, the viral envelope protein induces macrophage colony-stimulating factor (M-CSF). This pro-survival cytokine down regulates the TRAIL (tumor necrosis factor-related apoptosis-inducing ligands) receptor and up regulates the anti-apoptotic genes Bfl -1 and Mcl-1 enabling HIV to survive inside the macrophages. HIV invades the macrophage through CCR5 a chemokine receptor and through binding of gp120 to CD4 [ 73 ] . Macropinocytosis as a route of entry of HIV-1 into macrophages [ 74 ] also enables intracellular protection. Leshmania prevent activation of macrophages by inhibiting secretion of cytokines such as the infl ammatory response IL-1 and tumor necrosis factor beta (TNF-beta) or T-lymphocyte activation (IL-12) and produce various immunosuppressive signaling molecules, such as arachidonic acid metabolites and the cytokines TNFbeta and IL-10. L. chagasi induces TNF-beta production in the immediate environment of the infected human macrophage, and this may lead to inhibition of immune responses [ 75 ] . Further, this pathogen induces alteration of host cell signaling. Macrophages infected with L. donovani or L. mexicana have shown altered Ca 2+ dependent responses, such as chemotaxis and production of ROI [ 76 , 77 ] . Based on the adaptive mechanisms microorganisms reside in different cells and at different locations in the cells. Treating diseases therefore, necessitates an understanding of both the resident cells and target organelles. Illustrative examples of microorganism and their cellular/organelles targets are listed out in Table 3 .4 . The granulocytes are classifi ed as neutrophils, eosinophils, or basophils on the basis of cellular morphology. Neutrophils play the major role in the body's defence. Neutrophils are produced in the bone marrow by hematopoiesis. They are released into blood where they circulate for 7-10 h and migrate into tissues where they have a life span of a few days. During infection the bone marrow releases more than usual Tularemia -Cytosol Al-Khodor [ 91 ] number of neutrophils, which migrate to the site of the infection. They act by both oxygen-dependent and oxygen-independent pathways to kill microbes. Neutrophils exhibit a larger respiratory burst than macrophages and consequently are able to generate more reactive oxygen intermediates and reactive nitrogen intermediates. In addition, neutrophils express higher levels of defensins than macrophages do. Hence, neutrophils are more active than macrophages in killing ingested microorganisms. Dendritic cells are antigen-presenting cells and constitute 0.5-1 % of the leukocyte population in the peripheral blood mononuclear cells. They are found mostly in nonlymphoid tissues and organs such as skin, heart, liver, lungs, and mucosal surfaces. The function of these cells is to initiate, stimulate and regulate a T cell response which includes antigen-specifi c T lymphocytes, Th1/Th2 modulation, regulatory T cell induction and peripheral T cell deletion. There are four types of dendritic cells, i.e. Langerhans cells, myeloid dendritic cells, plasmacytoid dendritic cells and infi ltrating infl ammatory dendritic epidermal cells. CD1b, CD11a, CD11b and CD11c, the thrombospondin receptor (CD36), and the mannose receptor (CD206), present on infl ammatory dendritic epidermal cells, are known to be involved in the uptake of bacterial components. In case of Mycobacterium tuberculosis infection, alveolar macrophages (dust cells), along with dendritic cells engulf bacteria and exhibit innate as well as an adaptive immune response. Combined efforts by macrophages and dendritic cells establish protective immunity in 90 % of infected individuals. Natural killer cells (NKC) are non-phagocytic cells present mostly in mammalian and avian species [ 92 ] . NKC express surface receptors for the Fc portion of IgG and their function is to mediate antibody-dependent cytotoxicity against tumor target cells [ 93 ] . It is also suggested that NKC play a role in resistance against some microbial infections. NKC also play a role in natural genetic resistance to infections caused by cytomegalovirus and herpes simplex type I [ 94 , 95 ] . However, there is also evidence against the role of NKC in resistance to some other viruses [ 96 ] . Lymphocytes are cells present 99 % in the lymph and constitute 20-40 % of the body's white blood cells. There are approximately ~10 10 -10 12 lymphocytes in the human body, and this can vary with body weight and age. They circulate in the lymph and blood, and can migrate into tissue spaces and lymphoid organs, enabling integration with the immune system. The two main categories of lymphoid cells that can recognise and react against a wide range of specifi c antigens are B lymphocytes or B cells and T lymphocytes or T cells. The main function of B cells is to produce antibodies against antigens [ 97 ] . Each of the approximately 1. Natural T lymphocytes mature in the thymus region and survive in the periphery. The chief function of T cells is to respond to signals associated with tissue destruction and to minimise the collateral tissue damage they cause [ 98 ] . T cells express T-cell receptors (TCR) which are a composite of polypeptides including CD3 and either of one of the two membrane molecules, CD8 and CD4. TCR recognises virus infected cells and cancer cells. However, unlike B cells, TCR does not recognise free antigen, unless it is bound to MHC molecules on the membrane of antigen presenting cells. The main function of T cells is to induce death of virus infected cells by secretion of cytotoxins and cytokines which activates B cells, macrophages and cytotoxic T cells. T cells also play role in infectious diseases such as Leishmaniasis [ 99 ] , infection by hepatitis C virus (HCV), etc. Their ability to confi ne exuberant immune reactivity, associated with many chronic infections is benefi cial the host due to limited tissue damage [ 100 ] . Infectious diseases are also located in cells other than cells of the RES. Such cells include hepatocytes, epithelial cells and erythrocytes. Hepatocytes are located in the liver and are major site for infections such as hepatitis B/C and malaria. The hepatocytes are discussed in greater detail in Chapter 6 of this book. Epithelial cells bind together to form the epithelial tissue which is held together by adherens, tight junctions, gap junctions and desmosomes. The functions of epithelial cells are boundary and protection of vital organs, transportation, absorption, secretion, lubrication and movement. These epithelial cells can be readily attacked by microbes such as HIV virus, infl uenza, Herpes Simplex virus (HSV-2) and cause infections. Furthermore, erythrocytes are infected and act as hosts for plasmodium causing malaria, one of the current fatal infections posing serious challenges. The introduction of antimicrobial agents such as penicillin resulted in a major breakthrough to decrease morbidity and mortality caused by infectious diseases. Antibiotics represented one of the greatest discoveries. This euphoria was short lived due to adverse effects and the emergence of drug resistance. Conventional therapy when associated with side effects or necessitates long term treatment, results in low patient compliance. Further inadequate drug concentration within cells is a major barrier for effective treatment of intracellular diseases. Increasing the dose, however, resulted in enhanced toxicity. Mono-drug therapy evolved into multi-drug therapy, and enabled a good degree of success and continues to form standard therapy, even today. Classic examples include the multi-drug combination for tuberculosis AKT2, AKT3, AKT4 comprising 2, 3 or 4 drugs, respectively. The HAART combination for AIDS and two drug combinations for malaria are also examples of successful therapy. Nevertheless, the alarming rate at which drug resistance is occurring, and more so the emergence of multi-drug resistance are a matter of great concern. Tuberculosis is one such major disease which has evolved from Resistant to Multi-drug Resistant(MDR) to total drug resistant (TDR), the latest being extremely drug resistant tuberculosis (XXDR), wherein, resistance is seen to almost all known antitubercular drugs. The emergence of multi-drug resistance is attributed to a number of factors. Pathogens resort to different mechanisms to avoid intracellular killing. Some pathogens secrete exotoxins which destroy phagocytes and prevent phagocytosis. Bacteria with pore forming cytolysins avoid the phagosome and also escape lysosomal destruction [ 101 -105 ] . Certain bacteria interfere with the production of cytotoxic metabolites of phagocytes or contain the antioxidant proteins, thereby overcoming the effects of RNIs or ROIs and cause obstruction in phagocytosis [ 106 , 107 ] . Bacteria adhere to surfaces, aggregate and form a hydrated polymeric matrix comprising of exopolysaccharide known as biofi lms [ 108 ] . Biofi lms are developed by various bacteria such as Salmonella , Streptococcus, Vibrio cholerae, Klebsiella pneumonia and Haemophilus infl uenzae. Further some cells in the biofi lm experience nutrient limitation and therefore survive in the starved state. Such cells are slow growing cells and less susceptible to antimicrobial agents [ 109 ] . Certain cells in a biofi lm adapt a different and protected phenotype. Biofi lms are resistant to antibodies, phagocytes, and antibiotics . Although p hagocytes reach the biofi lms, they become frustrated and release their enzymes, which cause damage to the tissue around the biofi lm. Release of bacteria through the damaged biofi lm results in dissemination of the infection, leading to acute infection in the surrounding tissues [ 110 , 111 ] . Effl ux pump genes and proteins are present in almost all organisms. Effl ux pumps thwart the entry of an antibiotic in the bacterial cell and export an antibiotic from the cell. As effl ux pumps can be specifi c for one substrate or for drugs of dissimilar structure, they can be associated with multi-drug resistance. Multi-drug-resistance effl ux pumps are a known cause for the development of bacterial resistance against antibiotics. Bacterial effl ux-pump proteins related with MDR are divided into fi ve families namely the ATP binding cassette (ABC) superfamily, the major facilitator superfamily (MFS), the multi-drug and toxic-compound extrusion (MATE) family, the small multidrug resistance (SMR) family and the resistance nodulation division (RND) family [ 112 ] . Multi-drug resistance occurs, when effl ux proteins are overexpressed on the cell, and easily identify and effi ciently expel a broad range of antibiotics from the cells [ 113 ] . Gram-negative bacteria express several families of transporters which cause resistance [ 114 ] . Gram-positive bacteria mainly Staphylococcus aureus and Streptococcus pneumoniae express MDR effl ux pumps. S. aureus (responsible for skin and soft-tissue infections) overexpress MFS effl ux pump NorA which enables resistance to chloramphenicol and fl uoroquinolones. The S. pneumoniae MFS effl ux pumpPmrA exports the fl uoroquinolones ciprofl oxacin, norfl oxacin, and also expels the dyes acrifl avine and ethidium bromide [ 115 -117 ] Escherichia coli EmrE express a member of the small multi-drug resistance (SMR) superfamily and AcrAB-TolC, a member of the resistance-nodulation-cell division (RND) superfamily. Vibrio parahaemolyticus overexpress NorM, a member of the multi-drug and toxic compound extrusion (MATE) superfamily. Multi-drug-resistant tuberculosis (MDR-TB) is appearing as a ghost among the MDR bacteria because TB patients are at high risk of death due to failure of treatment. It is evident that MDR exhibits p55 effl ux pumps which play a crucial role in the pathogenicity of the microorganisms, and is responsible for the effl ux of tetracycline and aminoglycosides. This has opened a vast array for research in identifying mutants which are responsible for overexpressing these protein pumps in cases of elevated virulence [ 112 ] . Chemical modifi cation of antibiotics resulting in their inactivation and hence, ineffective dug concentration can be a cause of bacterial resistance. The inactivation reactions include hydrolysis, redox, and group transfer. Hydrolysis is the major cause of degradation of beta lactam antibiotics. The group transfer approach is the most varied and includes modifi cation by thiol transfer, glycosylation, acyl transfer, ribosylation, nucleotidylation and phosphorylation transfer. Drugs which are degraded by group transfer are aminoglycoside, chloramphenicol, rifamycin, macrolides, etc. [ 118 ] . One important strategy to overcome the limitation of conventional drug delivery is to deliver high therapeutic payloads intracellularly. This could ensure high efficacy, coupled with low toxicity to provide major advantages. Targeted nanocarriers provide high promise as potential drug delivery systems with the capacity to address this specifi c challenge. Targeted nanocarriers could therefore prove to be the magic wand. Passive and active targeting approaches could be relied on to achieve organ based targeting (fi rst order), specifi c cell based targeting in an organ (second order) and cell organelle based targeting (third order) [ 119 ] . A major requirement, however, besides reaching the targeting site is to ensure adequate concentration and adequate retention at the site. Passive targeting can be described as deposition of drug or drug-carrier systems at a particular location due to pharmacological or physicochemical factors [ 120 ] . Passive targeting can be achieved by exploiting pathophysiological and anatomical opportunities. Introduction of drugs directly into various anatomical sites for example lungs and the eye by using non-invasive or invasive methods such as catheters or direct injections can enable local targeting. These site specifi c drug delivery methods limit systemic toxicity of the drug thus reducing adverse effects of drugs in the nontarget tissues [ 121 ] . Exploiting altered pathological conditions in diseased tissues are strategies that can be adopted for passive targeting for example chemotactic factors released in infected or infl amed tissues increased permeability of vascular tissues, decreased pH and/or increased temperature [ 122 , 123 ] . Increased vascular permeability specifi cally in cancers has enabled passive targeting of nanocarriers and is cited as the enhanced permeation and retention effect (EPR) effect [ 124 ] . Surface properties such as particle size, shape, hydrophobicity and surface charge have great impact on macrophage activation and phagocytosis. Particle size plays essential role in distribution and elimination of nanocarriers [ 125 ] . Particles size can infl uence attachment, adhesion, phagocytosis, distribution, circulation half-life and endocytic pathways [ 126 , 127 ] . The opsonisation and phagocytosis of particles is strongly affected by size of nanocarriers. Although macrophages engulfed 0.2 versus 2 μm IgG-coated spherical particles by different mechanisms, they followed similar kinetics [clathrin endocytosis versus Fc-receptor mediated phagocytosis]. Phagocytic uptake is generally observed with polymeric particles and liposomes with high particle size [>200-microns] [ 128 ] . Table 3 .7 highlights the size of a number of nanocarriers evaluated for targeted delivery in infectious diseases. A broad range of non-spherical shaped particles studied including cylinders, cubes, hemispheres, ellipsoids, cones and complex shapes like fi lamentous, biconcave discoid showed varying effects on phagocytosis [ 169 ] . Non-spherical shaped particles bypassed phagocytosis due to incomplete actin structure formation. Particle shape affected attachment and internalisation during phagocytosis [ 170 ] . For instance oblate ellipsoids show best attachment and internalisation by phagocytosis, while prolate ellipsoids showed good attachment but poor internalisation. Champion et al. reported that worm-like particles showed low phagocytosis as compared to spherical particles of the same volume [ 169 ] . Asymmetric polymer lipid nanostructures (LIPOMER) of Doxycycline hydrochloride (DH) in the range of (250-400 nm) [ 171 ] revealed enhanced splenic delivery. The irregular shape of the LIPOMER coupled with rigidity resulted in fi ltration and non-phagocytic accumulation to reveal splenotropy in sinusoidal spleen models, rat, rabbit and dog. A high spleen liver ratio of 6.7:0.53 was seen in the dog model (Fig. 3.2 ) [ 172 ] . Surface properties like hydrophobicity and surface charge also impact opsonisation, phagocytosis and biodistribution of nanoparticles [ 173 ] . Hydrophobic nanocarriers are readily coated by complement proteins, albumin, and immunoglobulin and scavenged by RES [ 174 ] . Surface charge of particles also infl uences interaction and stability with cells [ 175 ] . Reports suggest that positively charged particles showed high phagocytic uptake over negatively charged particles probably due to better interaction with the negatively charged cell membrane. Cationic and neutral nanocarriers are less taken up by RES as compare to negatively charge [ 176 -178 ] . However, negatively charged nanoparticles can potentially attach to cationic sites on the macrophages namely the scavenger receptors, which facilitate their uptake by RES [ 179 ] . For details on infl uence of particle size, shape and charge readers are directed to the following reviews [ 126 , 180 ] . Active targeting, defi ned as specifi c targeting of drugs or drug containing nanocarriers by anchoring active agents or ligands, provides selectivity, recognisability and potential to interact with specifi c cells and tissues in the body [ 181 ] . Targeting by attaching ligands has been investigated as an additional strategy to enhance translocation of antimicrobials inside cells. Attaching ligands facilitates greater uptake and can be mediated by various mechanisms. The membrane of macrophages expresses various receptors to facilitate the internalisation of cargoes inside the cell and their degradation. Receptor mediated endocytosis (RME) permits the rapid internalisation of ligand attached particles as compared to untargeted particles [ 182 ] . The common RME mechanisms are macropinocytosis, clathrin dependent endocytosis (CDE), caveolae-mediated endocytosis and clathrin independent endocytosis (CIE). Each approach exhibits different binding and internalisation mechanisms. Further, the predominant uptake mechanism is often dictated by the nature of the ligand. Receptor mediated processes are relatively slower than phagocytic processes, with the ligand playing an important role. The sizes, geometry, charge and density of the ligand signifi cantly infl uences receptor mediated endocytosis [ 183 ] . For more references readers can refer to [ 182 , 184 , 185 ] . Table 3 .5 lists the endocytic pathways, endosome morphology and the proteins involved in the endocytic pathways. Macrophages possess large number of surface receptors which help in the process of recognition and endocytosis of engineered particulate carriers. Infection of macrophages leads to changes in the expression pattern of the concerned receptors, which can be exploited for targeted drug delivery employing nanocarriers. Table 3 .6 is a summary of the important receptors on macrophages and illustrative examples of ligands for the same that could play a role in designing targeted nanocarriers for infectious disease therapy. CD14 [ 213 ] , Decay accelerating factor (CD55), Endo180 [ 214 ] are also receptors which could be targeted. Nevertheless, ligands for the same need to be explored. All known nanocarriers can be effectively employed for targeted delivery in intracellular infections. Both passive and active targeting approaches have been evaluated. The following Tables 3.7 and 3.8 illustrate examples of nanocarriers, limited to major anti-infective agents for active and passive targeting, respectively. Size being a major parameter infl uencing targeting to RES. Table 3 .7 also highlights the size of nanocarriers, which is a primary factor in passive targeting. Tuberculosis is persistent and deadly infectious disease, caused Mycobacterium tuberculosis which is non-specifi cally phagocytosed by alveolar macrophages. Malaria is a complex disease caused by plasmodium and majorly resides in non-RES cells like red blood cells (RBCs) and hepatocytes. Entry of the parasite into the brain causes cerebral malaria. Malaria can be targeted at the exoerythrocytic stage by targeting RBCs, or targeting the hypnozoites to tackle malarial relapse and further in case of cerebral malaria targeting the brain. Increased permeability of infected RBCs is seen after 12-16 h of plasmodium invasion through formation of channels. These channels are "new permeability pathways" (NPPs) which allow entry of molecules such as dextran, protein A and IgG2a antibody thereby differentiating the non-infected and infected RBCs. Such pathways could be targeted to enable high drug loading in the erythrocytes specifi cally through design of nanocarriers of <80 nm [ 241 ] . This could be supported through design of stealth nanocarriers which could enable long circulation, using various stealth agents like poly(ethyleneglycol) (PEG), Pluronic, etc. [ 242 ] . Chloroquine liposomes anchored with anti-erythrocyte F (ab′)2 were studied for targeting to erythrocytes [ 243 ] . Hepatocytes the residence of hypnozoites expresses the asialoglycoprotein receptor (ASGPR), which is overexpressed in infections. Targeting this receptor using nanocarriers anchored with ASGPR ligands is a strategy for hepatocyte targeting. Joshi et al. prepared in situ primaquine nanocarboplex of primaquine phosphate anchored with pullulan as the ASGPR ligand for specifi c targeting to hepatocytes. Signifi cantly, enhanced hepatic accumulation with preferential accumulation in the hepatocytes and a high hepatocytes/nonparenchymal cells ratio of 75:25 confi rmed hepatocyte targeting [ 244 ] . Transferrin (Tf)-anchored solid lipid nanoparticles (SLNs) were intravenously administered for targeting quinine dihydrochloride to the brain, in cerebral malaria. Compared to conventional SLNs or drug solution the Tf-SLNs signifi cantly enhanced the brain uptake of quinine [ 234 ] . A major feature of HIV that complicates therapy is the existence of HIV in multiple reservoirs, which include various cellular and anatomical sites [ 245 ] . The typical reservoirs are the liver, spleen, lungs, GIT and genital tract with the brain and bone marrow representing remote sites [ 246 ] . Targeted delivery for HIV therefore needs to address delivery to maximum sites simultaneously to achieve remission. One strategy that we propose is a combination of nanocarriers of size <100 nm to target remote sites and size >200 nm target major RES organs (Unpublished data). Viral replication is inhibited by the antioxidant glutathione. Erythrocytes containing glutathione (GSH) in combination with azidothymidine (AZT) and didanosine (DDI) showed higher reduction in viral DNA in bone marrow and brain as compared to DDI + GSH alone [ 247 ] . Immunoliposomes containing siRNA for targeting the lymphocyte function-associated antigen-1 (LFA-1) integrin, which is expressed on all leukocytes, was selectively taken up by T cells and macrophages, the primary site of HIV. Further, in vivo administration of anti-CCR5 siRNA resulted in leukocytespecifi c gene silencing that was sustained for 10 days [ 248 ] . Nanogels comprising non-reverse transcriptase inhibitors (NRITs) decorated with a peptide for brain specifi c apolipoprotein E (apoE) receptors, showed tenfold suppression of retroviral activity and decrease infl ammation in humanised mouse model of HIV-1 infection in the brain [ 249 ] . Targeted drug delivery for the therapy of veterinary infections assumes immense importance not only for improved animal health but due to the challenges posed by zoonotic diseases. About 13 zoonotic diseases including brucellosis, tuberculosis, trypanosomiasis, cysticercosis and others are related to 2.4 billion cases of infection in humans and over two million deaths annually [ 166 , 167 ] . Such infections exist both in domestic animals and wild life. The close proximity of humans especially with such domestic animals is a cause of global concern. The WHO policy of "Cull and Kill" results not only in the loss of lives but also heavy monetary losses to the farmer. Targeted treatment strategies using nanodrug delivery systems could provide a revolutionary strategy to benefi t both the animals and man. The benefi ts of targeted nanomedicine strategies are slowly gaining recognition as evident from a number of reported studies. Liposomes have been used by many researchers for treating various veterinary diseases such as Leishmaniasis [ 250 , 251 ] , Brucellosis [ 252 ] , Blastomycosis [ 253 ] , Babesiosis [ 254 ] , etc. Patil et al. [ 171 ] developed an asymmetric lipomer. This is a combination of polymer-lipid containing doxycycline which could have application in the treatment of intracellular infections that are primarily resident in the spleen like brucellosis, ehrlichiosis, etc. A number of studies are reported on horses infected with babesiosis, Streptococcus equi, T. gondii and Strongylus vulgaris infections using liposomes [ 254 ] , polymeric nanospheres [ 255 ] , dendrimers [ 256 ] and micelles [ 257 ] respectively. A recent study revealed the improved therapy of theileiriosis in cattle with solid lipid nanoparticles (SLN) of buparvaquone [ 258 ] . SLN revealed comparable effect with the intramuscular injection at signifi cantly lower doses. Nanodrug delivery systems have also been evaluated in dogs, sheep and pigs. For details on nanodrug delivery applications in targeted delivery in veterinary infections, readers are directed to the following reference [ 259 ] . Targeted delivery for infectious diseases has immense scope. Tackling infections using nanodrug delivery systems could provide a practical alternative as a short term strategy. A rate-limiting factor however would be the serious concerns of toxicity. Nanodrug delivery systems due to their high intracellular delivery could precipitate new and unknown toxicities. Evolving strategies to predict the same is an important path forward. While vaccines could probably provide the ultimate cure and control, vaccine development is a complex process and not yet easily attained as evident from the limited success stories. However, designing nano-vaccines targeted to exhibit greater cellular response is also a near future prospect. Pentraxins: structure, function, and role in infl ammation Collectins -soluble proteins containing collagenous regions and lectin domains -and their roles in innate immunity Ficolins and infectious diseases Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations in Tlr4 gene Cutting edge: Toll-like receptor 4 (TLR4)-defi cient mice are hyporesponsive to lipopolysaccharide: evidence for TLR4 as the Lps gene product Endotoxin-tolerant mice have mutations in Toll-like receptor 4 (Tlr4) Recognition of double-stranded RNA and activation of NF-kappaB by Toll-like receptor 3 Differential roles of TLR2 and TLR4 in recognition of gram-negative and gram-positive bacterial cell wall components A Toll-like receptor recognizes bacterial DNA The innate immune response to bacterial fl agellin is mediated by Tolllike receptor Professional and non-professional phagocytes: an introduction Cell size of alveolar macrophages: an interspecies comparison Distinct role of macrophages in different tumor microenvironments Exploring the full spectrum of macrophage activation A microbial strategy to multiply in macrophages: the pregnant pause Mechanism of phagolysosome biogenesis block by viable Mycobacterium tuberculosis Yersinia pestis can reside in autophagosomes and avoid xenophagy in murine macrophages by preventing vacuole acidifi cation Lack of acidifi cation in Mycobacterium phagosomes produced by exclusion of the vesicular proton-ATPase Mycobacterium leprae surface components intervene in the early phagosome-lysosome fusion inhibition event Internalized Listeria monocytogenes modulates intracellular traffi cking and delays maturation of the phagosome Inhibition of macrophage phagosome-lysosome fusion by Salmonella typhimurium Inhibition of phagolysosomal biogenesis by the Leishmania lipophosphoglycan Avoidance of innate immune mechanisms by the protozoan parasite, Leishmania spp in protozoans in macrophages Phagosome acidifi cation blocked by intracellular Toxoplasma gondii Helicobacter pylori phagosome maturation in primary human macrophages Characterization of the catalytic subunit of Trypanosoma cruzi cyclic AMP-dependent protein kinase Trimming the fat: a Brucella abortus survival strategy Interaction of Leishmania parasites with dendritic cells and its functional consequences Association of Legionella pneumophila with the macrophage endoplasmic reticulum Two distinct defects in intracellular growth complemented by a single genetic locus in Legionella pneumophila Life on the inside: the intracellular lifestyle of cytosolic bacteria Survival of intracellular pathogens within macrophages Listeriolysin O: a genuine cytolysin optimized for an intracellular parasite Expression of the Rickettsia prowazekii pld or tlyC gene in Salmonella enterica serovar typhimurium mediates phagosomal escape Distinct isoforms of phospholipase A2 mediate the ability of Salmonella enterica serotype typhimurium and Shigella fl exneri to induce the transepithelial migration of neutrophils A potential new pathway for Staphylococcus aureus dissemination: the silent survival of S. aureus phagocytosed by human monocyte-derived macrophages Selective receptor blockade during phagocytosis does not alter the survival and growth of Mycobacterium tuberculosis in human macrophages Macrophage receptors for Mycobacterium tuberculosis Multicellular and aggregative behavior of Salmonella typhimurium strains is controlled by mutations in the agfD promoter Salmonella enterica serovar Typhimurium DT104 displays a rugose phenotype A novel relationship between O-antigen variation, matrix formation, and invasiveness of Salmonella enteritidis Thin aggregative fi mbriae and cellulose enhance long-term survival and persistence of Salmonella Thin, aggregative fi mbriae mediate binding of Salmonella enteritidis to fi bronectin The multicellular morphotypes of Salmonella typhimurium and Escherichia coli produce cellulose as the second component of the extracellular matrix Biofi lm formation by Escherichia coli O157:H7 on stainless steel: effect of exopolysaccharide and Curli production on its resistance to chlorine Effect of heat, acidifi cation, and chlorination on Salmonella enterica serovar Typhimurium cells in a biofi lm formed at the air-liquid interface Genetic analysis of Salmonella enteritidis biofi lm formation: critical role of cellulose Salmonella produces an O-antigen capsule regulated by AgfD and important for environmental persistence Lower intracellular hydrogen peroxide levels in cells overexpressing CuZn-superoxide dismutase Phenotypic switching of Cryptococcus neoformans occurs in vivo and infl uences the outcome of infection HIV-1 and the macrophages Human immunodefi ciency virus type 1 entry into macrophages mediated by macropinocytosis Activation of TGF-beta by Leishmania chagasi: importance for parasite survival in macrophages The effect of parasitization by Leishmania mexicana on macrophage function in vitro Subversion mechanisms by which immune response: a signaling point of leishmania parasites can escape the host Optimal cytoplasmic transport in viral infections Adaptation of the Brucellae to their intracellular niche Brucella evades macrophage killing via virb-dependent sustained interactions with the endoplasmic reticulum Differing infl uences of virus burden and immune activation on disease severity in secondary dengue-3 virus infections Membrane association of hepatitis C virus nonstructural proteins and identifi cation of the membrane alteration that harbors the viral replication complex Change of processing and nucleocytoplasmic transport of mRNA in HSV-1-infected cells Transgenic mice with intracellular immunity to infl uenza virus How the parasitic bacterium Legionella pneumophila modifi es its phagosome and transforms it into rough ER: implications for conversion of plasma membrane to the ER membrane An amphipathic sulphated glycoconjugate of Leishmania: characterization with monoclonal antibodies Assessing the contributions of the LiaS histidine kinase to the innate resistance of listeria monocytogenes to nisin, cephalosporins, and disinfectants The biochemistry of intracellular parasitism Innate immune response in the gut against Salmonella -review Adaptive changes in gene expression of Mycobacterium tuberculosis during the development of the infection Triggering Ras signalling by intracellular Francisella tularensis through recruitment of PKCa and bI to the SOS2/GrB2 complex is essential for bacterial proliferation in the cytosol Natural killer cells: their role in defenses against disease Natural killer cells may be the only cells in normal mouse lymphoid cell populations endowed with cytolytic ability for antibody-coated tumour target cells Genetic control of natural resistance to infection and malignancy Genetic infl uences on the augmentation of natural killer (NK) cells during murine cytomegalovirus infection: correlation with patterns of resistance Natural cell-mediated immunity during viral infections The cellular basis of the immune response. an approach to immunobiology Regulating the regulators Regulatory T cells: friend or foe in immunity to infection? Natural regulatory T cells in infectious disease Fusion of azurophil granules with phagosomes and activation of the tyrosin ekinase Hck are specifi cally inhibited during phagocytosis of mycobacteria by human neutrophils Legionella pneumophila DotA protein is required for early phagosome traffi cking decisions that occur within minutes of bacterial uptake Apoptosis in macrophages and alveolar epithelial cells during early stages of infection by Legionella pneumophila and its role in cytopathogenicity Legionella pneumophila utilizes the same genes to multiply within Acanthamoeba castellanii and human macrophages Survival of Mycobacterium avium and Mycobacterium tuberculosis in acidifi ed vacuoles of murine macrophages Interactions between professional phagocytes and Brucella spp Nitric oxide-mediated crosstalk between Helicobacter and gastric mucosa Bacterial biofi lms: a common cause of persistent infections Growth rate control of adherent bacterial populations The diffusion characteristics of antibiotics in mucus glycoprotein gels A review of experimental measurements of effective diffusive permeabilities and effective diffusion coeffi cients in biofi lms Multidrug-resistance effl ux pumps not just for resistance Multidrug effl ux pumps and resistance: regulation and evolution Antibiotic effl ux mechanisms The multidrug effl ux transporter of Bacillus subtilis is a structural and functional homolog of the Staphylococcus NorA protein Nucleotide sequence and characterization of the Staphylococcus aureus nor A gene, which confers resistance to quinolones Effl ux-mediated fl uoroquinolone resistance in Staphylococcus aureus Bacterial resistance to antibiotics: enzymatic degradation and modifi cation Drug targeting Targeted drug conjugates: principles and progress Nanosystems in drug targeting, opportunities and challenges Targeted drug delivery to enhance effi cacy and shorten treatment duration in disseminated Mycobacterium avium infection in mi host factors infl uencing the preferential localization of sterically stabilized liposomes in klebsiella pneumoniae-infected rat lung tissue Neutrophil transepithelial migration: evidence for sequential, contact-dependent signaling events and enhanced paracellular permeability independent of transjunctional migration Tumor vascular permeability and the EPR effect in macromolecular therapeutics: a review Characterization of the size, shape, and state of dispersion of nanoparticles for toxicological studies The effect of particle design on cellular internalization pathways Nanocarriers entry into the cell: relevance to drug delivery Antimicrobial effectiveness of liposomal polymyxin B against resistant Gram-negative bacterial strains vitro activities of free and liposomal drugs against Mycobacterium avium-M. intracellular complex and M. tuberculosis Biological evaluation of pyrazinamide liposomes for treatment of Mycobacterium tuberculosis Rifabutin encapsulated in liposomes exhibits increased therapeutic activity in a model of disseminated tuberculosis Liposome-entrapped ampicillin in the treatment of experimental murine listeriosis and salmonellosis Treatment of Brucella canis and Brucella abortus in vitro and in vivo by stable plurilamellar vesicle-encapsulated aminoglycosides Liposome-incorporated ciprofl oxacin in treatment of murine salmonellosis Parasitic diseases: liposomes and polymeric nanoparticles versus lipid nanoparticles Liposomes as carriers of the antiretroviral agent dideoxycytidine-5 %-triphosphate Gelatin nanocarriers as potential vectors for effective management of tuberculosis Development and characterization of guar gum nanoparticles for oral immunization against tuberculosis In vivo/in vitro pharmacokinetic and pharmacodynamic study of spray-dried poly-(dl-lactic-co-glycolic) acid nanoparticles encapsulating rifampicin and isoniazid Preparation and antibacterial activity evaluation of rifampicin-loaded poly lactide-co-glycolide nanoparticles Macrophage delivery of nanoformulated antiretroviral drug to the brain in a murine model of NeuroAIDS Targeted delivery of antibiotics to intracellular chlamydial infections using PLGA nanoparticles Targeted delivery of arjunglucoside I using surface hydrophilic and hydrophobic nanocarriers to combat experimental leishmaniasis Increase in gentamicin uptake by cultured mouse peritoneal macrophages and rat hepatocytes by its binding to polybutylcyanoacrylate nanoparticles Targeted delivery of nanoparticles for the treatment of lung diseases Encapsulation of moxifl oxacin within poly(butyl cyanoacrylate) nanoparticles enhances effi cacy against intracellular Mycobacterium tuberculosis Nanoencapsulation increases quinine antimalarial effi cacy against Plasmodium berghei in vivo In vitro antileishmanial activity of amphotericin B loaded in poly(ε-caprolactone) nanospheres Inhalable microparticles containing isoniazid and rifabutin target macrophages and "stimulate the phagocyte" to achieve high effi cacy Microparticle-based lung delivery of INH decreases INH metabolism and targets alveolar macrophages Lopinavir loaded solid lipid nanoparticles (SLN) for intestinal lymphatic targeting Transmucosal transport of tobramycin incorporated in solid lipid nanoparticles after duodenal administration to rats. Part ii tissue distribution Solid lipid nanoparticles as drug carriers Incorporation and retention of the lipophilic prodrug 3´-azido-3´-deoxythymidine palmitate Solid lipid nanoparticles enhance the delivery of the HIV protease inhibitor, atazanavir, by a human brain endothelial cell line Pharmacokinetics, tissue distribution and relative bioavailability of isoniazid-solid lipid nanoparticles Targeted intracellular delivery of antituberculosis drugs to mycobacterium tuberculosis-infected macrophages via functionalized mesoporous silica nanoparticles Niosomal delivery of isoniazid development and characterization Formulation, antimalarial activity and biodistribution of oral lipid nanoemulsion of primaquine A novel injectable water-soluble amphotericin B-arabinogalactan conjugate Targeting potential and anti-HIV activity of lamivudine loaded mannosylated poly (propyleneimine) dendrimer Glycodendrimeric nanoparticulate carriers of primaquine phosphate for liver targeting Targeted killing of Leishmania donovani in vivo and in vitro with amphotericin B attached to functionalized carbon nanotubes Characterisation of copper oxide nanoparticles for antimicrobial applications Investigations into the antibacterial behavior of copper nanoparticles against Escherichia coli Global trends in emerging infectious diseases Antibacterial activity of ZnO nanoparticle suspensions on a broad spectrum of microorganisms Effect of iron oxide and gold nanoparticles on bacterial growth leading towards biological application Role of target geometry in phagocytosis Polymer particle shape independently infl uences binding and internalization by macrophages Role of lipids in enhancing splenic uptake of polymer-lipid (LIPOMER) nanoparticles Particle shape: a new design parameter for passive targeting in splenotropic drug delivery Particle size, surface coating, and PEGylation infl uence the biodistribution of quantum dots in living mice PLGA nanoparticles of different surface properties: preparation and evaluation of their body distribution Effect of surface properties on nanoparticle-cell interactions Surface-modifi ed biodegradable albumin nano-and microspheres. II: effect of surface charges on in vitro phagocytosis and biodistribution in rats Drug targeting by surface cationization The effect of surface charge on in vivo biodistribution of PEG-oligocholic acid based micellar nanoparticles The class B scavenger receptors SR-BI and CD36 are receptors for anionic phospholipids Targeting to macrophages: role of physicochemical properties of particulate carriers liposomes and microspheres on the phagocytosis by macrophages Lymphatic targeting with nanoparticulate system Endocytic mechanisms for targeted drug delivery The effect of nanoparticle size, shape, and surface chemistry on biological systems Mechanisms of endocytosis Endocytosis and intracellular transport of nanoparticles: present knowledge and need for future studies Simultaneous binding of PtdIns P2 and clathrin by AP180 in the nucleation of clathrin lattices on membranes Yolk protein uptake in the oocyte of the mosquito Aedes aegypti The multiple faces of caveolae Caveolin, a protein component of caveolae membrane coats Caveolins: structure and function in signal transduction Flotillin-1 defi nes a clathrin-independent endocytic pathway in mammalian cells Coassembly of fl otillins induces formation of membrane microdomains, membrane curvature, and vesicle budding Arf family GTP loading is activated by, and generates, positive membrane curvature Characterization of a nonclathrin endocytic pathway: membrane cargo and lipid requirements Clathrin-independent endocytosis: a unique platform for cell signaling and PM remodelling Clathrin-independent endocytosis: new insights into caveolae and noncaveolar lipid raft carriers Quantitative microscopy reveals 3D organization and kinetics of endocytosis in rat hepatocytes A dynamin-cortactin-Arp2/3 complex mediates actin reorganization in growth factor-stimulated cells Clathrinindependent endocytosis used by the IL-2 receptor is regulated by Rac1, Pak1 and Pak2 Interleukin 2 receptors and detergent-resistant membrane domains defi ne a clathrin-independent endocytic pathway Uptake of Pneumocystis carinii mediated by the macrophage mannose receptor Tuftsin-bearing liposomes in treatment of macrophage-based infections Tuftsin (an Ig-associated tetrapeptide) triggers the immunogenic function of macrophages: implications for activation of programmed cells Microglial scavenger receptors and their roles in the pathogenesis of Alzheimer's disease Targeting the hemoglobin scavenger receptor CD163 in macrophages highly increases the anti-infl ammatory potency of dexamethasone The function of Fcγ receptors in dendritic cells and macrophages Primary structure of the mannose receptor contains multiple motifs resembling carbohydrate-recognition domains Folate receptor β is expressed by tumor-associated macrophages and constitutes a marker for M2 anti infl ammatory/regulatory macrophages Folate receptor β as a potential delivery route for novel folate antagonists to macrophages in the synovial tissue of rheumatoid arthritis patients Targeted drug delivery via the transferrin receptor-mediated endocytosis pathway The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors Macrophage complement receptors and pathogen clearance IFN-lambdas mediate antiviral protection through a distinct class II cytokine receptor complex Toll receptors, CD14, and macrophage activation and deactivation by LPS Drug delivery system: targeting of pentamidines to specifi c sites using sugar grafted liposomes Effi cient drug delivery to alveolar macrophages and lung epithelial lining fl uid following pulmonary administration of liposomal ciprofl oxacin in rats with pneumonia and estimation of its antibacterial effects Hyaluronan-modifi ed and regular multilamellar liposomes provide sub-cellular targeting to macrophages, without eliciting a pro-infl ammatory response The role of apolipoprotein E in Alzheimer's disease Targeting Leishmania (L.) chagasi amastigotes through macrophage scavenger receptors: the use of drugs entrapped in liposomes containing phosphatidylserine Therapeutic effi cacies of isoniazid and rifampin encapsulated in lung-specifi c stealth liposomes against Mycobacterium tuberculosis infection induced in mice Aerosolized liposome-based delivery of amphotericin B to alveolar macrophages Uptake and processing of immunoglobulin-coated liposomes by subpopulations of rat liver macrophages Sterically stabilized liposomes bearing anti-HLA-DR antibodies for targeting the primary cellular reservoirs of HIV-1 Targeted delivery of indinavir to HIV-1 primary reservoirs with immunoliposomes Development and optimization of curcumin-loaded mannosylated chitosan nanoparticles using response surface methodology in the treatment of visceral leishmaniasis Ionic complexation as a noncovalent approach for the design of folate anchored rifampicin gantrez nanoparticles Surfacemodifi ed cobalt oxide nanoparticles: new opportunities for anti-cancer drug development TAT-conjugated nanoparticles for the CNS delivery of anti-HIV drugs Targeted brain delivery of AZT via transferrin anchored pegylated albumin nanoparticles Transferrin-and transferrin-receptor-antibody-modifi ed nanoparticles enable drug delivery across the blood-brain barrier (BBB) Mannan-coated gelatin nanoparticles for sustained and targeted delivery of didanosine: in vitro and in vivo evaluation Mannan-modifi ed solid lipid nanoparticles for targeted gene delivery to alveolar macrophages Mannosylated nanoparticulate carriers of rifabutin for alveolar targeting Transferrin-conjugated solid lipid nanoparticles for enhanced delivery of quinine dihydrochloride to the brain Polyamidoamine dendrimers-capped carbon dots/Au nanocrystal nanocomposites and its application for electrochemical immunosensor Toxicological investigation of surface engineered fi fth generation poly (propyleneimine) dendrimers in vivo Targeting of efavirenz loaded tuftsin conjugated poly (propyleneimine) dendrimers to HIV infected macrophages in vitro Intracellular macrophage uptake of rifampicin loaded mannosylated dendrimers Macrophages targeting of amphotericin B through mannosylated multiwalled carbon nanotubes Inhalable microparticles containing nitric oxide donors: saying NO to intracellular mycobacterium tuberculosis Protein transport across the parasitophorous vacuole of Plasmodium falciparum: into the great wide open Design aspects of poly(alkylcyanoacrylate) nanoparticles for drug delivery Functional drug targeting to erythrocytes in vivo using antibody bearing liposomes as drug vehicles Receptor-mediated hepatocyte-targeted delivery of primaquine phosphate nanocarboplex using a carbohydrate ligand The challenge of viral reservoirs in hiv-1 infection HIV disease management in the highly active antiretroviral therapy (HAART) era New drug combinations for the treatment of murine AIDS and macrophage protection RNAi-mediated CCR5 silencing by LFA-1-targeted nanoparticles prevents HIV infection in BLT mice Nano-NRTIs demonstrate low neurotoxicity and high antiviral activity against HIV infection in the brain Effi cacy of the liposome trifl uralin in the treatment of experimental canine leishmaniasis Reduced tissue parasitic load and infectivity to sand fl ies in dogs naturally infected by Leishmania (Leishmania) chagasi following treatment with a liposome formulation of meglumine antimoniate Effi cacy of various treatment regimens, using liposomal streptomycin in cows with brucellosis Use of an amphotericin B lipid complex for treatment of blastomycosis in dogs Liposomal diamidine (imidocarb), preparation and animal studies Enhanced mucosal immunoglobulin A response and solid protection against foot-and-mouth disease virus challenge induced by a novel dendrimeric peptide Evaluation of immune responses in sheep induced by DNA immunization with genes encoding GRA1, GRA4, GRA6 and GRA7 antigens of Toxoplasma gondii Effi cacy of ivermectin in injectable and oral paste formulations against eight-week-old Strongylus vulgaris larvae in ponies Buparvaquone loaded solid lipid nanoparticles for targeted delivery in theleriosis Targeted nanomedicine strategies for livestock infections. Nanotechnology for Animal Health and Production