throbber
Multifunctional, stimuli-sensitive
`nanoparticulate systems for
`drug delivery
`
`Vladimir P. Torchilin1,2
`
`Abstract | The use of nanoparticulate pharmaceutical drug delivery systems (NDDSs)
`to enhance the in vivo effectiveness of drugs is now well established. The development
`of multifunctional and stimulus-sensitive NDDSs is an active area of current research.
`Such NDDSs can have long circulation times, target the site of the disease and enhance
`the intracellular delivery of a drug. This type of NDDS can also respond to local stimuli
`that are characteristic of the pathological site by, for example, releasing an entrapped
`drug or shedding a protective coating, thus facilitating the interaction between
`drug-loaded nanocarriers and target cells or tissues. In addition, imaging contrast
`moieties can be attached to these carriers to track their real-time biodistribution and
`accumulation in target cells or tissues. Here, I highlight recent developments with
`multifunctional and stimuli-sensitive NDDSs and their therapeutic potential for diseases
`including cancer, cardiovascular diseases and infectious diseases.
`
`Enhanced permeability
`and retention (EPR) effect
`The property through which
`macromolecules (such as
`nanoparticles) accumulate
`in areas of inflammation
`including tumours, owing to
`the increased vascular
`permeability or abnormal
`blood vessel architecture.
`
`1Center for Pharmaceutical
`Biotechnology and
`Nanomedicine, Northeastern
`University, 140 The Fenway,
`Room 214, 360 Huntington
`Avenue, Boston,
`Massachusetts 02115, USA.
`2Faculty of Pharmacy,
`King Abdulaziz University,
`Jeddah 21589, Saudi Arabia.
`e‑mail: v.torchilin@neu.edu
`doi:10.1038/nrd4333
`Published online
`7 October 2014
`
`Nanoparticulate pharmaceutical drug delivery systems
`(NDDSs) are widely used in pharmaceutical research
`and in clinical settings to enhance the effectiveness
`of diagnostic agents and drugs, including anticancer,
`antimicrobial and antiviral drugs1,2. The types of nano­
`carriers that exist are diverse and include the following:
`liposomes; polymeric nanoparticles; polymeric micelles;
`silica, gold, silver and other metal nanoparticles; carbon
`nanotubes; solid lipid nanoparticles; niosomes; and dendri­
`mers. The use of NDDSs can overcome several problems
`that are associated with traditional drugs, such as poor
`aqueous solubility, low bioavailability and nonspecific
`distribution in the body.
`The first generation of NDDSs mainly aimed to
`address single challenges, such as the need to increase
`drug stability in vivo and the circulation time in the
`blood, or the need to target a drug to a specific tissue or
`pathology. Now, research has led to the development of
`NDDSs that can perform two or more functions (either
`simultaneously or sequentially) to overcome multiple
`physiological barriers to optimize delivery and deliver
`their loads (which can be single or multiple) to the
`required target sites (such as organs, tissues, cells) or
`specific pathologies in the body3 (FIG. 1). The properties
`of multifunctional NDDSs include the ability to bear
`a sufficient load of a drug or DNA­related material, have
`
`increased circulation times (through the use of soluble
`polymers) and target the intended site of action both
`nonspecifically (for example, via the enhanced permea­
`bility and retention (EPR) effect) and specifically (via the
`attachment of target­specific ligands). In addition,
`multifunctional NDDSs can respond to several stimuli
`that are characteristic of the pathological site, which
`is achieved through the inclusion of components that
`react to abnormal pH, temperature and redox condi­
`tions, and to the overexpression of certain biological
`molecules. Multifunctional NDDSs can also respond
`to stimuli from outside the body, such as magnetic or
`ultrasound fields, and can be supplemented with an
`imaging contrast moiety to enable their biodistribution,
`target accumulation or the efficacy of the therapy to be
`monitored.
`Although as yet there is no broadly recognized and
`accepted single classification system for multifunctional
`NDDSs, they can generally be divided into three groups.
`The first group consists of drug­loaded NDDSs that com­
`bine at least two different functions, such as longevity,
`targetability, stimuli­sensitivity or cell penetration. The
`second group of NDDSs, in addition to the previously
`described properties, are loaded with more than one drug
`and/or gene therapy­related material, such as antisense
`oligonucleotides or small interfering RNAs (siRNAs).
`
`NATURE REVIEWS | DRUG DISCOVERY
`
` VOLUME 13 | NOVEMBER 2014 | 813
`
`© 2014 Macmillan Publishers Limited. All rights reserved
`
`REVIEWS
`
`

`

`Multifunctional, stimuli-sensitive
`nanoparticulate systems for
`drug delivery
`
`Vladimir P. Torchilin1,2
`
`Abstract | The use of nanoparticulate pharmaceutical drug delivery systems (NDDSs)
`to enhance the in vivo effectiveness of drugs is now well established. The development
`of multifunctional and stimulus-sensitive NDDSs is an active area of current research.
`Such NDDSs can have long circulation times, target the site of the disease and enhance
`the intracellular delivery of a drug. This type of NDDS can also respond to local stimuli
`that are characteristic of the pathological site by, for example, releasing an entrapped
`drug or shedding a protective coating, thus facilitating the interaction between
`drug-loaded nanocarriers and target cells or tissues. In addition, imaging contrast
`moieties can be attached to these carriers to track their real-time biodistribution and
`accumulation in target cells or tissues. Here, I highlight recent developments with
`multifunctional and stimuli-sensitive NDDSs and their therapeutic potential for diseases
`including cancer, cardiovascular diseases and infectious diseases.
`
`Enhanced permeability
`and retention (EPR) effect
`The property through which
`macromolecules (such as
`nanoparticles) accumulate
`in areas of inflammation
`including tumours, owing to
`the increased vascular
`permeability or abnormal
`blood vessel architecture.
`
`1Center for Pharmaceutical
`Biotechnology and
`Nanomedicine, Northeastern
`University, 140 The Fenway,
`Room 214, 360 Huntington
`Avenue, Boston,
`Massachusetts 02115, USA.
`2Faculty of Pharmacy,
`King Abdulaziz University,
`Jeddah 21589, Saudi Arabia.
`e‑mail: v.torchilin@neu.edu
`doi:10.1038/nrd4333
`Published online
`7 October 2014
`
`Nanoparticulate pharmaceutical drug delivery systems
`(NDDSs) are widely used in pharmaceutical research
`and in clinical settings to enhance the effectiveness
`of diagnostic agents and drugs, including anticancer,
`antimicrobial and antiviral drugs1,2. The types of nano­
`carriers that exist are diverse and include the following:
`liposomes; polymeric nanoparticles; polymeric micelles;
`silica, gold, silver and other metal nanoparticles; carbon
`nanotubes; solid lipid nanoparticles; niosomes; and dendri­
`mers. The use of NDDSs can overcome several problems
`that are associated with traditional drugs, such as poor
`aqueous solubility, low bioavailability and nonspecific
`distribution in the body.
`The first generation of NDDSs mainly aimed to
`address single challenges, such as the need to increase
`drug stability in vivo and the circulation time in the
`blood, or the need to target a drug to a specific tissue or
`pathology. Now, research has led to the development of
`NDDSs that can perform two or more functions (either
`simultaneously or sequentially) to overcome multiple
`physiological barriers to optimize delivery and deliver
`their loads (which can be single or multiple) to the
`required target sites (such as organs, tissues, cells) or
`specific pathologies in the body3 (FIG. 1). The properties
`of multifunctional NDDSs include the ability to bear
`a sufficient load of a drug or DNA­related material, have
`
`increased circulation times (through the use of soluble
`polymers) and target the intended site of action both
`nonspecifically (for example, via the enhanced permea­
`bility and retention (EPR) effect) and specifically (via the
`attachment of target­specific ligands). In addition,
`multifunctional NDDSs can respond to several stimuli
`that are characteristic of the pathological site, which
`is achieved through the inclusion of components that
`react to abnormal pH, temperature and redox condi­
`tions, and to the overexpression of certain biological
`molecules. Multifunctional NDDSs can also respond
`to stimuli from outside the body, such as magnetic or
`ultrasound fields, and can be supplemented with an
`imaging contrast moiety to enable their biodistribution,
`target accumulation or the efficacy of the therapy to be
`monitored.
`Although as yet there is no broadly recognized and
`accepted single classification system for multifunctional
`NDDSs, they can generally be divided into three groups.
`The first group consists of drug­loaded NDDSs that com­
`bine at least two different functions, such as longevity,
`targetability, stimuli­sensitivity or cell penetration. The
`second group of NDDSs, in addition to the previously
`described properties, are loaded with more than one drug
`and/or gene therapy­related material, such as antisense
`oligonucleotides or small interfering RNAs (siRNAs).
`
`NATURE REVIEWS | DRUG DISCOVERY
`
` VOLUME 13 | NOVEMBER 2014 | 813
`
`© 2014 Macmillan Publishers Limited. All rights reserved
`
`REVIEWS
`
`

`

`Liposome, micelle
`Drug A
`Drug B
`
`Moiety sensitive to:
`• pH
`• Temperature
`• Redox
`• Enzyme activity
`
`Polymer coat, such
`as PEG, to increase
`longevity
`
`Targeting agent:
`• Antibodies
`• Transferrin
`• Peptides
`
`Cell-penetrating
`peptide such as
`HIV TAT peptide
`
`Imaging or
`contrast agent:
`• Gd
`• 64Cu
`
`Figure 1 | Schematic of a drug-loaded, multifunctional, stimuli-sensitive NDDS.
`Drugs (Drug A and Drug B) can be loaded into a pharmaceutical nanocarrier, such as
`Nature Reviews | Drug Discovery
`a liposome or polymeric micelle. Depending on the purpose of the nanoparticulate
`pharmaceutical drug delivery system (NDDS), various agents can be added to the
`nanoparticle to target the NDDS to a particular tissue, to increase cell penetration,
`to enable imaging or to release the drugs in response to a given stimulus.
`PEG, poly(ethylene glycol).
`
`The third group consists of so­called theranostic NDDSs,
`which have an additional diagnostic label for use with
`current clinical imaging modalities.
`Research in the area of multifunctional NDDSs4,5 is
`very active, but substantial work remains to make them
`a clinical reality. Here, I highlight recent developments
`relating to multifunctional NDDSs. The majority of the
`currently available data relate to cancer, although there
`are some examples with other diseases.
`
`NDDS longevity and targeting
`One of the most common uses of NDDSs is to com­
`bine prolonged circulation times with targetabilty. Such
`NDDSs are particularly useful for tumour targeting
`because tumours (as well as other inflammation zones)
`usually have increased vascular permeability as well as
`poor lymphatic drainage6,7. This enables long­circulating
`NDDSs to accumulate in tumours through the EPR
`effect, which forms the basis for passive targeting8.
`Never the less, EPR­based drug delivery strategies face
`several challenges. First, tumours — especially large,
`solid tumours — are pathophysiologically heterogene­
`ous. Some parts of such tumours are not vascularized,
`do not exhibit the EPR effect, may have sizeable necrotic
`areas9,10 and have varied microvascular permeability10.
`In addition, the increased interstitial pressure that exists
`within tumours may limit the EPR­mediated accumula­
`tion of NDDSs even if the vasculature is leaky11.
`NDDSs that are used for passive targeting and/or
`spontaneous accumulation must have long circulation
`times to ensure that sufficient drug is delivered to the tar­
`get tissue. The usual approach to obtain long­circulating
`NDDSs is to coat them with hydrophilic and flexible
`polymers, such as poly(ethylene glycol) (PEG), as was
`first suggested for liposomes12. The pegylation of NDDSs
`prevents their interaction with opsonins and impedes
`their capture and clearance by the mononuclear phago­
`cyte system. However, pegylated NDDSs can induce the
`production of antibodies that can accelerate the blood
`clearance of nanoparticles (as was demonstrated for
`
`Passive targeting
`The mechanism through
`which nanoparticulate
`pharmaceutical drug delivery
`systems tend to accumulate
`in tumours, probably through
`the enhanced permeability
`and retention effect.
`
`Quantum dots
`Nanometre­scale particles of
`semiconductor materials that
`have quantum mechanical
`properties.
`
`Active targeting
`The mechanism through which
`specific moieties attached to
`nanoparticulate pharmaceutical
`drug delivery systems force
`them to interact with a specific
`type of cell or tissue.
`
`clinically used pegylated liposomes), particularly after
`repetitive administration; this is the so­called accelerated
`blood clearance phenomenon13,14.
`Although PEG is still the gold­standard polymer
`that is used to prepare long­circulating NDDSs, other
`hydrophilic polymers that are used include poly[N­
`(2­hydroxypropyl) methacrylamide]15, poly(acryloyl
`morpholine), poly­N­vinylpyrrolidones16 and polyvinyl
`alcohol17. In general, the concept of pegylation, as well as
`the use of alternative polymers for longevity, is well
`established and reviewed; thus, in the next section only
`some of the key developments in this area are briefly
`mentioned. Importantly, the shape of NDDSs can also
`influence their pharmacokinetics and biodistribution18,19.
`Liposomal long­circulating NDDSs are the most
`frequently studied type of NDDS; however, synthetic
`amphiphilic polymers have also been used to sterically
`stabilize several other types of NDDS to alter their bio­
`distribution. For example, pegylation of gold nanoparti­
`cles reduced their uptake by the mono nuclear phagocyte
`system and their subsequent clearance from the body20.
`One potential application of pegylated gold nanoparticles
`is their use in photothermal tumour therapy (known as
`ablation) following their accumulation in the tumour21.
`Pegylated methotrexate­conjugated poly­l­lysine
`dendrimers also accumulated efficiently in solid tumours
`in rats and mice via the EPR affect22, and pegylation
`reduced the toxicity of positive charge­bearing dendri­
`mers in cell­culture experiments23. Quantum dots can also
`be modified by pegylation. In mice, pegylated quantum
`dots had prolonged circulation times, which were attrib­
`utable to their decreased uptake in organs (such as the
`spleen and liver) in which the mononuclear phagocyte
`system is active24.
`Active targeting of NDDSs can be achieved by attaching
`targeting ligands, such as monoclonal antibodies, trans­
`ferrin, various peptides, folate, aptamers (single­stranded
`oligonucleotides) or certain sugar moieties, onto their
`surface25,26. To prevent steric hindrance between the tar­
`geting moiety and the protective polymer (such as PEG),
`the targeting ligand is usually attached to the chemically
`activated distal end of the NDDS­grafted polymeric
`chain27. An example of such active targeting of NDDSs
`is pegylated doxorubicin­loaded liposomes that have
`human epidermal receptor 2 (HER2; also known as
`ERBB2)­specific antibodies attached; these were suc­
`cessfully used to target HER2­overexpressing SK­BR3
`cells in mice28. A nucleosome­specific monoclonal
`antibody (mAb 2C5) that recognized multiple types of
`tumour cells via tumour cell­surface­bound nucleosomes
`improved the targeting of doxorubicin­loaded liposomes
`to tumour cells and increased cancer cell cytotoxicity in
`in vitro and in vivo models29, including nude mice xeno­
`grafted with the U­87 cell line, which was derived from
`a human glioblastoma30. In another example of active
`targeting, pegylated gold nanoparticles conjugated to
`monoclonal F19 antibodies were used as targeted label­
`ling agents for human pancreatic carcinoma tissues31.
`Lactoferrin­conjugated PEG–polylactic­acid nanoparti­
`cles improved the delivery of the conjugated particles to
`the brain in an experimental murine model32.
`
`814 | NOVEMBER 2014 | VOLUME 13
`
` www.nature.com/reviews/drugdisc
`
`© 2014 Macmillan Publishers Limited. All rights reserved
`
`R E V I E W S
`
`

`

`An interesting approach to increase targeting centres
`on self­assembling polyalkylcyanoacrylate­based nano­
`particles. These types of nanoparticles could serve as
`pharmaceutical carriers for various drugs and could be
`produced in pegylated and ‘activated’ forms, enabling
`various ligands to be easily attached33. Polyisohexyl­
`cyanoacrylate nanoparticles loaded with doxorubicin
`produced by BioAlliance Pharma as Livatag (doxoru­
`bicin Transdrug) are currently in a Phase III trial in
`Europe and in the United States as a second­line treat­
`ment of hepatocellular carcinoma after sorafenib, at a
`stage with no available approved treatment (Livatag
`press release; see Further information).
`Aptamers that impart affinity and specificity by elec­
`trostatic, hydrogen or hydrophobic bonding, but not via
`the base pairing, have also been used as stable and effi­
`cient targeting moieties for NDDSs. For example, lipo­
`somal NDDSs modified with specific aptamers have
`been used to target leukaemia cells34.
`Targetability can also be applied to stimuli­sensitive
`NDDSs. Gemcitabine­loaded pegylated pH­sensitive
`liposomes modified with an epidermal growth factor
`receptor (EGFR)­specific antibody efficiently inhib­
`ited tumour growth in mice35. Micellar NDDSs that
`were composed of amphiphilic conjugates of PEG and a
`co­polymer of poly(ε­caprolactone) and poly(malic acid)
`loaded with doxorubicin conjugated to poly(malic acid)
`via a pH­sensitive bond were modified with folate for
`enhanced cellular uptake36. This preparation provided an
`enhanced release of doxorubicin at lowered pH values
`inside cancer cells36. NDDSs with similar properties
`based on pH­sensitive polymethacrylate (PMA)­grafted
`poly(amidoamine) nanocarriers and additionally
`pegylated and modified with a folate moiety efficiently
`delivered paclitaxel to tumours and inhibited tumour
`growth in mice37.
`Clearly, the separation of long­circulating NDDSs into
`those that achieve active targeting and passive targeting is
`conditional. That is, both phenomena are closely connected
`because NDDSs will accumulate in the target area via the
`EPR effect (passive targeting) before ligand­mediated
`interaction with target cells (active targeting) occurs.
`Another interesting issue is associated with the use of
`targeting ligands that are also naturally present (in their
`free form) in the circulation, such as folate, transferrin or
`certain peptides. Although one may expect competition
`between the ligands attached to NDDSs and the native
`ligands for the binding sites, the success of folate­ and
`transferrin­targeted NDDSs (see below) demonstrates
`that this potential problem can be overcome. This is
`probably achieved through multipoint interactions of
`ligand­modified NDDSs with the target and/or because
`of the rapid recirculation of the cognate receptors, which
`provides sufficient opportunities for an interaction of the
`NDDS with the receptor.
`
`Disease applications for NDDS-based therapies
`Cardiovascular diseases. NDDSs hold promise as thera­
`peutic, diagnostic and theranostic agents for cardio­
`vascular diseases38, particularly atherosclerosis39 (FIG. 2).
`Multifunctional micellar NDDSs that combined a
`
`targeting pentapeptide, a fluorophore and a drug targeting
`atherosclerotic plaques by specifically binding to clotted
`plasma proteins were used to visualize atherosclerotic
`lesions in a mouse model. This preparation was shown
`to increase the amount of the anticoagulant agent biva­
`lirudin that was delivered to the lesion40. Because the
`increased uptake of low­density lipoproteins (LDLs)
`by resident macrophages in plaques is associated with
`the progression of atherosclerosis, targeting plaque­
`associated macrophages could be a method of inhibit­
`ing LDL uptake41. Indeed, targeted polymeric NDDSs
`loaded with pravastatin specifically and dramatically
`inhibited the phagocytic activity of macrophages without
`affecting non­target cells42.
`Multimodal diagnostic micellar NDDSs that were
`fluorescent and paramagnetic and targeted to macro­
`phage scavenger receptors using specific antibodies have
`been used to visualize lesions in the abdominal aortas
`of mice with atherosclerosis43. Furthermore, polymeric
`NDDSs made of polyketals that were used to deliver
`small­molecule drugs, such as the p38 mitogen­activated
`protein kinase (MAPK) inhibitor SB239063, and anti­
`oxidant proteins, such as superoxide dismutase, to the area
`of the myocardial infarction in a rat model significantly
`improved cardiac function44.
`Liposome­based systems have also been frequently
`used to study the potential use of NDDSs in cardiovascular
`diseases45. For example, liposomes loaded with ATP or
`co­enzyme Q accumulated well in the infarcted areas of
`the myocardium and improved cardiac parameters in
`rat and rabbit models of myocardial infarct46,47. In a rat
`model of heart transplant, organ rejection was simulta­
`neously imaged and treated using multifunctional poly­
`meric NDDSs co­loaded with superparamagnetic iron
`oxide nanoparticles (SPIONs) and plasmid DNA48. The
`SPION acted as a magnetic resonance imaging (MRI)
`contrast agent, whereas the plasmid DNA suppressed the
`local immune response48. NDDSs for targeted thromb­
`o lytic therapy were developed by conjugating a throm­
`bolytic agent (recombinant tissue plasminogen activator)
`to dextran­coated iron oxide nanoparticles, which were
`additionally modified with a thrombus­targeted pep­
`tide that was sensitive to activated factor XIII49. These
`NDDSs had increased binding to the margins of intravas­
`cular thrombi and good thrombolytic activity in a mouse
`model of pulmonary embolism.
`The translation of these NDDSs and others identified
`in similar studies into clinical practice remains the primary
`challenge to advancing the field and will be addressed in
`the closing section of this article.
`
`Infectious diseases. NDDSs have a growing role in com­
`batting infectious diseases50. Of particular interest here
`are multicomponent NDDSs that contain metals, which
`can facilitate the formation of reactive oxygen species
`to eliminate pathogenic bacteria (FIG. 2). For example,
`NDDSs that combined iodinated chitosan and silver
`nanoparticles bound to and killed bacteria better than
`either component alone51. Hydrogel and glass­based
`nanoparticles capable of releasing nitric oxide facili­
`tated the cleaning and healing of open wounds in mice52.
`
`NATURE REVIEWS | DRUG DISCOVERY
`
` VOLUME 13 | NOVEMBER 2014 | 815
`
`© 2014 Macmillan Publishers Limited. All rights reserved
`
`R E V I E W S
`
`

`

`a Cardiovascular disease
`
`Atherosclerotic
`plaque
`
`Heart
`
`b Infectious disease
`
`Clot
`proteins
`
`NDDS
`
`Macrophage
`
`Clot
`proteins
`
`ROS
`
`Protein synthesis
`
`Bacterium
`
`Metals
`
`Antibiotics
`
`Figure 2 | Multifunctional and stimuli-sensitive NDDSs in cardiovascular pathologies and infectious diseases.
`Nature Reviews | Drug Discovery
`a | Nanoparticulate pharmaceutical drug delivery systems (NDDSs) can be engineered to target activated macrophages
`in atherosclerotic plaques or proteins present in a clot. These nanoparticles can thereby reduce plaque size through
`these two distinct mechanisms. b | NDDSs can be used to deliver metals to bacteria, where they can generate reactive
`oxygen species (ROS) to induce membrane blebbing and DNA damage. Antibiotics can also be delivered intracellularly
`using NDDSs.
`
`NDDSs can be used to deliver antibiotics inside cells;
`murine salmonellosis was successfully treated (with
`a greater than tenfold decrease in the level of pathogen
`in the liver and spleen of experimental mice) using gen­
`tamicin in silica xerogel­based NDDSs53. The bacteria­
`mediated degradation of a polyphosphoester core of
`antibiotic­loaded NDDSs by bacterial enzymes54 pro­
`vided an interesting example of a targeted, responsive
`NDDS. Nevertheless, this area is in early stages of devel­
`opment and more animal­based studies will be required
`to reveal its full potential.
`
`Cancer. The majority of preclinical research with NDDSs
`relates to cancer. Based on one of the most popular
`pharmaceutical nanocarriers — liposomes — several
`anticancer NDDSs are available at different stages of
`development (see TABLE 1 for a selected list).
`The next step in the development of anticancer NDDSs
`is to target them to tumours. Cancer cells contain sev­
`eral targets discussed below that can be used by NDDSs.
`Each of these targets could be specific for a certain type
`or types of cancer. Although it is possible that there will
`be overlapping expression of a target in cancer cells and
`normal cells, the level of expression is usually much higher
`in cancer cells; this characteristic is a prerequisite for an
`NDDS­targeting ligand.
`Folate has been used as a targeting ligand in pre­
`clinical studies of NDDSs, owing to its easy conjugation
`to nanocarriers, high affinity for folate receptors and the
`lower expression of folate receptors in normal cells then
`
`in cancer cells and activated macrophages that are typical
`of inflammatory diseases55. Numerous folate­ targeted
`liposomal systems for cancer have been described
`(reviewed in REFS 56,57). For example, liposomes modi­
`fied with folate via a PEG spacer efficiently delivered
`their cargo intracellularly through receptor­mediated
`endocytosis (which could bypass multidrug resistance),
`and doxorubicin­mediated cytotoxicity against cancer
`cells expressing folate receptors was 85­fold higher with
`drug­loaded targeted liposomes than with unmodified
`liposomes58. In addition, when glutathione­conjugated
`folate was coupled to a PEG–distearoylphosphatidyl­
`ethanolamine (PEG–DSPE) polymer and incorporated
`into the liposomal membrane, there was increased activity
`of the liposomal vincristine and favourable pharmaco­
`kinetics in mice59. Folate­based modified NDDSs loaded
`with paclitaxel60 or carboplatin61 had high antitumour
`activity in mouse models. Liposome targeting to folate
`receptors has also been used to improve gene delivery of
`the herpes simplex virus type 1 thymidine kinase suicide
`gene; the liposome preparation inhibited tumour growth
`in a mouse model of oral cancer62.
`Transferrin, an 80 kDa serum glycoprotein, binds
`to the transferrin receptor and is taken into cells via
`receptor­mediated endocytosis. Compared with normal
`cells, the expression of transferrin receptor in certain
`cancer cells is much higher because of their increased
`demand for iron. Consequently, transferrin­receptor­
`targeted NDDSs could be used for anticancer therapy.
`Indeed, this was demonstrated with doxorubicin­loaded
`
`816 | NOVEMBER 2014 | VOLUME 13
`
` www.nature.com/reviews/drugdisc
`
`© 2014 Macmillan Publishers Limited. All rights reserved
`
`R E V I E W S
`
`

`

`Table 1 | Liposome-based drugs for cancer therapy
`Drug formulation
`Indication
`Liposomal doxorubicin (Doxil, also known
`AIDS-related Kaposi’s sarcoma, recurrent
`as Caelyx; Janssen)
`ovarian cancer, metastatic breast cancer
`and multiple myeloma
`Metastatic breast cancer
`
`Status*
`Approved
`
`Approved
`
`Approved
`
`Phase III, active: NCT00617981
`Phase I/II, recruiting: NCT00826085
`
`Phase III, recruiting: NCT02112656
`Approved in China
`
`Phase II, completed: NCT01537536,
`NCT00448305
`Phase II, completed: NCT00377936
`Phase II, completed: NCT00542048
`Phase II, completed: NCT01190982
`
`Approved
`
`Approved
`
`Kaposi’s sarcoma, breast and ovarian
`cancer
`Hepatocellular carcinoma
`Breast cancer
`
`Hepatocellular carcinoma
`Solid tumours
`
`Breast cancer
`
`Pancreatic cancer
`Liver cancer
`Breast cancer
`
`Kaposi’s sarcoma
`
`Acute lymphoblastic leukaemia
`
`Non-pegylated liposomal doxorubicin
`(Myocet; Sopherion Therapeutics)
`Liposomal doxorubicin (LipoDox;
`Sun Pharma Global FZE)
`Thermally sensitive liposomal
`doxorubicin (Thermodox; Celsion )
`
`Liposomal paclitaxel (Lipusu; Luye
`Pharma)
`Liposomal paclitaxel (EndoTAG-1;
`Medigene)
`
`Liposomal paclitaxel (LEP-ETU;
`NeoPharm)
`Liposomal daunorubicin (DaunoXome;
`Galen)
`Liposomal vincristine (Marqibo; Talon
`Therapeutics)
`Liposomal cisplatin (LiPlaCis; LiPlasome
`Pharma)
`Liposomal cisplatin (Lipoplatin; Regulon)
`
`Liposomal cisplatin (SPI-077; Azla
`Corporation)
`Liposomal formulation of SN-38,
`the active metabolite of irinotecan
`(LE-SN38; NeoPharm)
`Liposomal irinotecan HCl: floxuridine
`mixture (CPX-1; Celator Pharmaceuticals)
`Liposomal irinotecan (MM-398;
`Merrimack Pharmaceuticals)
`Liposomal L-annamycin
`
`Refs
`165
`
`168,169
`
`170,171
`
`182–184
`
`166
`
`175,176
`
`202
`
`167
`
`172–174
`
`203
`
`175,
`177–179
`175,180,
`181
`175,185
`
`182
`
`204
`
`186,187
`
`188
`
`Advanced or refractory solid tumours
`
`Phase I, recruiting: NCT01861496
`
`Pancreatic cancer, non-small-cell lung cancer,
`head and neck cancer and breast cancers
`Ovarian cancer
`
`Phase I/II/III
`
`Phase II, completed: NCT00004083
`
`Colorectal carcinoma
`
`Phase II, completed: NCT00311610
`
`Colorectal cancer
`
`Phase II, completed: NCT00361842
`
`Metastatic pancreatic cancer
`
`Phase III, active: NCT01494506
`
`Acute lymphocytic leukaemia,
`doxorubicin-resistant blood cancer
`Hodgkin’s disease, non-Hodgkin’s lymphoma
`
`Phase I/II
`
`Phase I, active: NCT00364676
`
`Liposomal vinorelbine, INX-0125
`(AlocrestTM; Hana Biosciences)
`*ClinicalTrials.gov identifiers provided where appropriate.
`
`long­circulating transferrin­modified liposomes in a rat
`model of liver cancer63. Other examples of the anticancer
`effects of transferrin­targeted liposomes in mouse mod­
`els include those loaded with boron compounds64,65 and
`with ceramide66.
`EGFRs are frequently overexpressed in solid tum­
`ours67,68 and are therefore popular targets for NDDSs.
`HER2, which is overexpressed in approximately 20% of
`breast cancers68 and some other cancers, is also a popular
`target for NDDSs. For example, there was greater accu­
`mulation of HER2­targeted pegylated liposomes loaded
`with doxorubicin than of non­targeted liposomes in
`HER2­overexpressing breast cancer cells69 and in ascitic
`lymphoma cells70.
`
`The tumour vasculature is also an attractive target;
`destruction of the vasculature inhibits tumour growth
`and metastasis, it is not tumour type­specific and,
`importantly, NDDSs do not need to diffuse into the
`tumour mass to reach this target71. Targets that have
`been the focus of most studies include vascular endothe­
`lial growth factor (VEGF; especially for anti­angiogenic
`therapy), vascular cell adhesion molecule (VCAM),
`matrix metalloproteinases (MMPs) and integrins72,73.
`For example, peptides that bind specifically to the
`tumour vasculature coupled to liposomes loaded with
`doxorubicin improved drug efficacy against several
`human cancers in severe combined immunodeficient
`mice74. Pegylated VCAM­targeted immunoliposomes
`
`NATURE REVIEWS | DRUG DISCOVERY
`
` VOLUME 13 | NOVEMBER 2014 | 817
`
`© 2014 Macmillan Publishers Limited. All rights reserved
`
`R E V I E W S
`
`

`

`bound selectively to activated endothelial cells in vitro
`and accumulated in tumour vessels in vivo75. MMPs are
`overexpressed on the newly formed vessels and tumour
`tissues, and are involved in tissue remodelling, tumour
`invasiveness, resistance to apoptosis and metastasis. An
`antigen­binding fragment of the antibody against MMP
`that was conjugated to doxorubicin­loaded liposomes
`via a PEG spacer showed enhanced tumoural uptake and
`inhibited tumour growth in a mouse model76.
`
`Considerations for NDDS receptor targeting. The success
`of receptor­mediated targeting depends strongly on the
`target affinity and the density of the receptors that are
`present on the cell surface. In addition, with pegylated
`NDDSs, the surface­attached PEG could prevent or hinder
`the interaction between the targeting ligand and its recep­
`tor. In a sense, this limits the opportunities of targeting
`because not all potential targets are expressed at sufficient
`levels. It is also important to consider, on a case by case
`basis, whether spontaneous targeting (passive accumula­
`tion; based on the EPR effect) or ligand­mediated specific
`targeting (which clearly requires more synthetic effort)
`is preferable. In general, if good accumulation of a drug
`in the pathological tissue is the primary goal, then EPR­
`mediated accumulation may suffice. However, if the pres­
`ence of the drug inside the cell is needed then ligands that
`can be internalized may be required.
`
`Stimuli-sensitive NDDSs
`The use of NDDSs that respond to different types of
`

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket