improving drug delivery by use of lipid self-assembly par ticle

Upload: reza-ul-jalil

Post on 04-Apr-2018

215 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/30/2019 Improving Drug Delivery by Use of Lipid Self-Assembly Par Ticle

    1/4

    Professor Fredrik Tiberg was

    appointed President and CEO of

    Camurus in January 2003. He is

    also Professor of Surface and

    Colloid Chemistry at Lund

    University, Sweden. During 2001

    and 2002, he was Visiting Professor

    at Oxford University and Special

    Supernumerary Fellow of University

    College. Prior to his current

    position as President and

    CEO of Camurus, Professor Tiberg

    held a position as Vice President,

    Head of R&D at Camurus. He was

    appointed Associate Professor inPhysical Chemistry in 1998 and

    Professor in 1999. In 1996 he

    became Section Manager at the

    Institute for Surface Chemistry,

    Stockholm, Sweden. In these

    professional roles, Professor Tiberg

    has worked as consultant, scientific

    advisor, and project coordinator,

    serving a number of companies

    from small to multinational

    concerns. During the course of his

    distinguished academic career,

    Professor Tiberg has published more

    than 80 original scientific papers

    and co-authored several books. In

    1994 he received his PhD in

    Physical Chemistry from

    Lund University.

    a report by

    P r o f e s s o r F r e d r i k T i b e r g

    Professor of Surface and Colloid Chemistry, Lund University

    I n t r odu c t i o n

    Lipid-based drug delivery systems (DDS) can play a

    direct role in improving efficacy and drug safety,

    whereby new and improved therapies are possible.

    This has stimulated the creative design and study of

    lipid-based carriers for delivery through differentportals of the body. Colloidal-sized lipid particles in

    aqueous media, and their pre-concentrated dosage

    forms, have found important applications in a range

    of drug products, featuring all routes of

    administration. Lipid self-assembly structures even

    find use as matrices for injectable depot products.

    When dissolved in aqueous media, lipids self-

    assemble to form a wide range of mesophase

    structures (Figure 1). The complex architectures can

    be advantageously utilised to facilitate solubilisation,

    stabilisation, and delivery of different drugsubstances. Well-known self-assembly structures

    creatively utilised in pharmaceutical products include

    Sandimmun Neoral (microemulsion, Novartis),

    Diprivan (emulsion, AstraZeneca), and Doxil

    (Stealth liposome, ALZA).

    More recently, lipid-based, self-assembled liquid

    crystalline (LC) structures of cubic and hexagonal

    phases, and their corresponding nanoparticles

    (Cubosome and Hexosome), have emerged as

    potential carriers for several applications. The broad

    solubilisation spectrum together with high drugpayloads, as well as the ability to protect sensitive

    substances like peptides and proteins, and facilitate

    absorption, make these LC systems interesting

    alternatives to the more commonly used micelle,

    microemulsion, emulsion and liposome systems.

    Similar to microemulsions, emulsion and liposomes,

    they can be designed to self-disperse into colloidal

    particles. This property is essential in many

    applications where water-free liquid or powder pre-

    concentrates are the desired dosage forms.

    Lipids are ubiquitously distributed and playfundamental roles in the architecture and

    functionality of all living cells. Human health is

    dependent on the regular ingestion of various lipids

    because of their role in maintaining normal cellular,

    physiological and developmental function. Our

    natural delivery system operates via synergetic

    interplay between lipids and various enzymes. A

    wide variety of lipid self-assembly structures with

    specific structural and functional attributes can be

    prepared by exploiting the self-assembly properties

    unique to different lipids. Here, we briefly introducelipid-based systems used in pharmaceutical products

    and development.

    As noted above, lipid-based DDS represent a diverse

    group of formulations, each having different

    structural and functional characteristics which can be

    modified by adjusting the contents of different lipid

    excipients and other additives. The spectrum of

    varying properties seems unlimited, but is, in

    practice, restricted by cost, convenience, safety and

    regulatory demands. The introduction of new

    excipient combinations and DDS will most likelystimulate the use of lipid-based DDS and lead to new

    and improved therapies in the future. Many drugs on

    the market, and in different development stages, have

    non-optimal properties that potentially could be

    improved by a suitable delivery system (see Table 1).

    Here, we present different lipid-based DDS, illustrating

    their principal uses and potential future applications.

    Mi c e l l e s , M i c r o emu l s i o n s , a nd

    Emu l s i o n s

    These aggregates are the simplest self-assembly

    structures of lipids and surfactants with a widespread

    use in pharmaceutical products. The issue most

    frequently addressed by these systems is solubility. A

    low solubility and associated sluggish dissolution

    severely reduces the bioavailability of many water-

    insoluble drugs. Ideally, a formulation should enable

    uptake by keeping the drug in the dissolved state as a

    solubilisate throughout its GIT-transit. Successful oral

    lipid formulations on the market include Kaletra

    (lopinavir and ritonavir, Abbott) and Fortovase

    (saquinavir, Roche). Particularly efficient are self-emulsifying drug delivery systems, SEDDS. A

    hallmark of SEDDS is emulsification to a fine

    emulsion when released into the lumen, despite very

    gentle agitation. From a physicochemical point of

    Improv ing Drug De l i ve ry by use o f L ip id Se l f -a ssembly Part i c l e

    S t ruc ture s Beyond L iposomes and Emuls ions

    62

    Drug Delivery

    B U S I N E S S B R I E F I N G : P H A R M A O U T S O U R C I N G 2 0 0 5

  • 7/30/2019 Improving Drug Delivery by Use of Lipid Self-Assembly Par Ticle

    2/4

    Improving Drug Del ivery by use of Lip id Se l f -as sembly Par t i c le S t ruc tures

    view, the process is a phase inversion phenomenon,

    and the composition of a SEDDS must mediate very

    low interfacial tension between the oil and aqueous

    phases. The product Sandimmun Neoral

    (cyclosporin A, Novartis) uses this approach and is

    now the gold standard for treatment against rejection

    following organ transplant. In this respect it should be

    emphasised that the SEDDS microemulsions typically

    cannot be utilised for parenteral drug delivery because

    of safety issues regarding the excipients. This is due to

    the comparably high activity of the excipients

    employed in SEDDS formulations. On the other

    hand, micelles and o/w emulsions have traditionally

    been used extensively in parenteral formulations,

    using relatively mild surfactants or lipids, such as

    polysorbates, cremophors, egg lecithin, and soybean

    oil. Examples of drugs intended for the i.v. route,

    utilising micelles and o/w emulsions as solubilisation

    aids, include Taxol

    (paclitaxel, Bristol-MyersSquibb), which takes advantage of cremophor

    micelles, and Diprivan (propofol, AstraSeneca),

    which is an o/w emulsion.

    L i po somes

    Liposomes have been studied as drug carriers since the

    1970s, but it took about two decades to develop the

    first successful product applications. The constantly

    growing stream of liposome publications illustrates the

    enormous research effort going into this field. Based

    on present clinical applications of liposomes, the future

    is regarded with optimism, although developments

    have not yet lived up to earlier high expectations.

    Liposomes represent dispersed, closed, bilayer

    assemblies of the lamellar phase in excess water.

    Biologically active molecules, such as proteins, canbe incorporated into bilayers or the aqueous interior

    of the liposomes whereby primitive mimetics of

    biological cells are created. More importantly in this

    context, is that loading and encapsulation of drug

    substances in the interior aqueous core, or in the

    bilayer domain, is possible. Liposome characteristics

    depend on manufacturing protocol and excipient

    composition. They can be made in sizes ranging

    from unilamellar vesicles with a size as small as 20nm

    to multilamellar structures 10m in diameter.

    Table 1: Drug Delivery Issues Addressed by

    Lipid-based DDS

    1. Low aqueous solubility

    2. Storage andin vivo stabilities

    3. Local irritancy

    4. Poor absorption

    5. Rapid clearance

    6. Dose-limiting toxicities

    7. Poor distribution to target organs

    /NTIMEONTARGETDELIVERY

    )NNOVATIVENANOSCALEDRUGDELIVERYSYSTEMS

    WWWCAMURUSSE

    7EDONTWANTINNOVATIONWEWANT RESULTS

    7ENEEDPROFESSIONALBUSINESSPARTNERSNOTABUNCHOFACADEMICS

    7EHAVEADRUGDEVELOPMENTSCHEDULEWERE TRYING

    TOMEET #AN#AMURUSREALLYDELIVER

    7EVEHEARDITALLBEFOREAFTERALLWEVEBEENINTHISBUSINESSFORWELLOVERADECADE

    !NDWITHOVERPATENTSANDPRIORITYAPPLICATIONSVEONGOINGINDUSTRYCOLLABORATIONSANDTHREEUPCOMINGCLINICALTRIALSINSTEADOFGIVINGYOUASALESTALKLETUSACTUALLYSHOWYOUWHATOURLIQUIDCRYSTALTECHNOLOGIESCANDO

  • 7/30/2019 Improving Drug Delivery by Use of Lipid Self-Assembly Par Ticle

    3/4

    Among common lipid drug-carrier structures, the

    liposome structures are uniquely adjusted for

    delivery of hydrophilic drugs. This is due to the

    presence of an internal aqueous milieu encapsulated

    by an outer impermeable bilayer structure, whichacts as a chemical, mechanical, and electrical barrier

    against the ambient media. Due to low permeability

    of protons and other cations, they can be prepared

    with acidic internal pH, which facilitates active

    loading of water-soluble drugs (weak bases), and

    can be used to enhance drug stability. Mechanical

    properties and permeability of bilayer structures are

    determined by choice of components. Saturated

    phospholipids tend to be rigid and relatively

    impermeable, whereas unsaturated lipids are

    relatively flexible and more permeable. Current

    liposome drugs on the market, and in latedevelopment phase, are typically i.v. products

    featuring water-soluble and/or highly toxic/tissue-

    irritant active substances, like doxorubicin,

    daunorubicin and vincristine, and lipid-soluble

    amphotericin B. Benefits of i.v. liposome carriers

    include controlling the drug activity and toxicity.

    Inside the liposome the product is inactive.

    Avoidance of extravasation decreases the

    distribution volume and can increase the circulation

    half-life. The plasma half-life can be further

    enhanced by tethering stabilising polymer chains at

    the liposome surface, e.g. PEG-lipids. Indirectly,this leads to changes in the biodistribution: to tissues

    with enhanced vascular permeability to and,

    thereby, to passive targeting of regions of

    infection or inflammation, or to leaky blood vessels

    characteristic of angiogenesis. Aside from passive

    targeting, liposome structures can be equipped with

    homing devices, such as surface-tethered

    antibodies. Such liposomes become increasingly

    complex. It is noteworthy that even simple things

    are difficult in the pharmaceutical world, and that

    such complex systems will require a substantial

    upside in therapeutic index and market potential toeven be considered for drug development.

    Utilisation of liposomes in administration routes

    other than i.v. is, at present, limited because of the

    complexity of liposome formulations and the

    existence of better and simpler alternatives. There are

    a number of liposomal DDS on the market,

    however, that are intended for the s.c. or i.m. routes.

    One example of a depot technology based onliposomes is the DepoFoam from SkyePharma.

    Another current interesting clinical development

    concerns transdermal liposome drug products. Such

    systems are in clinical development, featuring actives,

    e.g. Transfersomes with ketoprofen developed by

    IDEA, claiming improved bioavailability.

    Nanopa r t i c l e s o f N on - l ame l l a r

    Ph a s e s Cub i c , H ex a gon a l

    and S pon ge

    As seen in Figure 1, lipids can self-assemble into

    different non-lamellar geometries. From a drug

    delivery perspective, these have attractive structural

    attributes and functional properties. In analogy with

    the liposomal dispersions, non-lamellar particles can

    be produced in multiphase systems, comprising a

    non-lamellar phase and excess water. This was first

    observed in a study of fat digestion, where cubic-

    phase particles were identified. It was later found that

    similarly structured particles could be prepared by

    dispersing glycerol monoleate (GMO) in water in the

    presence of surfactant and polymeric stabilisers. Thefull potential of non-lamellar colloidal particles,

    however, has not been realised in pharmaceutical

    applications. As with liposomes, drug delivery

    applications have been hampered due to the absence

    of suitable manufacturing methods for preparing

    structurally well-defined and stable dispersions, and

    because of the fact that only a few known lipid

    systems exhibit suitable phase behaviour. Recent

    developments allow scientists to take advantage of

    the true potential of non-lamellar phase structures in

    drug delivery applications.

    The main issues addressed by the non-lamellar LC

    phases, and their corresponding nanoparticles, are

    solubilisation, drug payload, and protection of

    sensitive drugs in vivo against rapid degradation by

    B U S I N E S S B R I E F I N G : P H A R M A O U T S O U R C I N G 2 0 0 5

    64

    Drug Delivery

    Figure 1: Different Self-assembly Objects Formed by Lipids/surfactants in Aqueous Media

    Oil in water

    Reversed

    micelles (L2)

    Reversed

    hexagonal (H11)Micelles (L1) Hexagonal (H1) Lamellar (L) Cubic (Q)

    Mirror plane Water in oil

    In this report, special emphasis is put on the water-in-oil-type of structures, such as the bicontinous cubic (Q) and the reversed hexagonal (HII) phas e.

  • 7/30/2019 Improving Drug Delivery by Use of Lipid Self-Assembly Par Ticle

    4/4

    Improving Drug Del ivery by use of Lip id Se l f -as sembly Par t i c le S t ruc tures

    endogenous enzymes. Examples of lipid-based

    liquid crystalline nanoparticles (LCNP) are shown

    in Figure 2. Especially noteworthy is the nano-

    structured interior of the particles, featuring both

    hydrophilic (aqueous) and lipophilic (lipid)

    domains. In contrast to liposomes, which consist

    mostly of water even for relatively small liposomeradii, the lipid content of typical LCNP is high

    (normally ranging 5070wt%). Because of

    coexisting hydrophilic and lipophilic domains, and

    the enormous surface area possessed by these

    structures, they have a broad spectrum of

    applicability, comprising lipophilic and amphiphilic

    bioactive agents, and peptide and protein drugs.

    In this context it can be pointed out that normal o/w

    emulsions are best adapted for strongly lipophilic

    drugs that require an oil medium for optimal

    solubilisation. Contrastingly, amphiphilic drugs, andin particular peptides and proteins, are often less

    compatible with emulsion systems. Also, compared

    with liposomes, drug payloads can be increased

    considerably using a LCNP DDS. An example is

    given in Figure 3, where loading capacity of

    cyclosporine A into LCNP carriers (Cubosome and

    Hexosome) is compared with that of liposome and

    microemulsion formulations, respectively.

    Important recent developments allow for the LCNP

    to be formed, by self-dispersion or self-

    emulsification, from a water-free formulationpreconcentrate similar to the SEDDS microemul-

    sions. The Flexosome particles in Figure 2 are based

    on a so-called sponge phase and can be produced

    simply by gentle magnetic stirring. High drug-

    payload characteristics of a broad spectrum of drugs,

    together with in situ emulsification, imply that the

    Flexosome system is ideal for oral applications

    where self-emulsifying properties are desired for

    convenience and optimal performance.

    Early non-lamellar LCNP preparations featured

    lipids that were not well-suited for parenteral drugdelivery, as exemplified by the unsaturated

    monoglyceride (uMG) GMO. Together with

    unreliable and scalable manufacturing schemes, the

    scope of application of the LCNP has been limited.

    Later improvements concerning both manufacturing

    schemes and excipient base have now finally

    facilitated the use of non-lamellar phases and

    nanoparticles in parenteral drug delivery. Using

    biodegradable and well-tolerated lipid excipients and

    reproducible, reliable, and scalable manufacturing

    schemes, the final obstacles for the use of non-

    lamellar LCNP DDS in parenteral applications hasnow been overcome.

    Other promising delivery routes of non-lamellar

    LCNP are the nasal and transdermal routes. The

    Eurocine L3 adjuvant is a sprayable nasal vaccine

    based on a low viscous sponge phase. Composed of

    endogenous and pharmaceutically accepted lipids, it

    is approved for human use and a successful phase I

    trial of nasal immunisation with diphtheria vaccine

    has recently been performed.

    Summary and F u tu r e

    Ou t l o ok

    Self-assembled LC structures provide a wide

    spectrum of structural and functional features. It

    appears that these attributes can be advantageously

    utilised to fit virtually any drug delivery challenge,

    making lipid-based DDS a successful approach.

    Key challenges related to manufacturing, stability,

    and reproducibility have been overcome, adding

    new opportunities of optimising drug product

    performance with regards to therapeutic index and

    patient convenience and compliance. Currently,

    non-lamellar LCNP are being implemented aspromising approaches in a number of late-stage

    development products. A growing pipeline of

    products utilising lipid-based LC and LCNP DDS

    can be foreseen in the near future.

    B U S I N E S S B R I E F I N G : P H A R M A O U T S O U R C I N G 2 0 0 5

    65

    Figure 2: Cryogenic Transmission Electron Microscopy (cryo-TEM) Images of

    Liquid Crystal Nanoparticles (LCNP). From Left to Right: Cubosome,

    Hexosome, and Flexoso me

    Figure 3: Drug Loading Capacity of Cubosome (QS)

    and Hexosome (HS) in Comparison with a

    Liposome (LS) and a Microemulsion (ME)

    Formulation. Cyclosporine A was used as a Model

    Drug and the Drug Loading Capacity is given in

    Weight of Drug per Excipient Weight

    30

    25

    20

    15

    10

    5

    0QS1 QS2 HS1 LS ME

    Carr

    iercapacity

    %