You are on page 1of 14

Journal of Controlled Release 190 (2014) 15–28

Contents lists available at ScienceDirect

Journal of Controlled Release


journal homepage: www.elsevier.com/locate/jconrel

Perspective Review

An overview of clinical and commercial impact of drug delivery systems


Aaron C. Anselmo, Samir Mitragotri ⁎
Department of Chemical Engineering, Center for Bioengineering, University of California, Santa Barbara 93106, USA

a r t i c l e i n f o a b s t r a c t

Article history: Drug delivery systems are widely researched and developed to improve the delivery of pharmaceutical com-
Received 13 February 2014 pounds and molecules. The last few decades have seen a marked growth of the field fueled by increased number
Accepted 28 March 2014 of researchers, research funding, venture capital and the number of start-ups. Collectively, the growth has led to
Available online 18 April 2014
novel systems that make use of micro/nano-particles, transdermal patches, inhalers, drug reservoir implants and
antibody–drug conjugates. While the increased research activity is clearly an indication of proliferation of the
Keywords:
Drug delivery
field, clinical and commercial translation of early-stage research ideas is critically important for future growth
Historical and interest in the field. Here, we will highlight some of the examples of novel drug delivery systems that
Perspective have undergone such translation. Specifically, we will discuss the developments, advantages, limitations and
Pharmaceutics lessons learned from: (i) microparticle-based depot formulations, (ii) nanoparticle-based cancer drugs,
Clinical translation (iii) transdermal systems, (iv) oral drug delivery systems, (v) pulmonary drug delivery, (vi) implants and
Lab to clinic (vii) antibody–drug conjugates. These systems have impacted treatment of many prevalent diseases including
diabetes, cancer and cardiovascular diseases, among others. At the same time, these systems are integral and
enabling components of products that collectively generate annual revenues exceeding US $100 billion. These
examples provide strong evidence of the clinical and commercial impact of drug delivery systems.
© 2014 Elsevier B.V. All rights reserved.

1. Introduction these abilities, many do. These limitations have given rise to substantial
research focused on the development of novel DDS.
Drug delivery systems (DDS) improve the administration and effica- Research in drug delivery has focused not only on improving oral
cy of pharmaceutical compounds including antibodies, peptides, and injectable systems, but also on opening additional routes of admin-
vaccines, drugs and enzymes, among others. Oral pills and injections istration including pulmonary [3], transdermal [4], ocular [5] and nasal
represent the most common mode of administering drugs today. A routes [6]. Each route has its own advantages and limitations (Table 1).
majority of small molecule drugs are delivered by pills. Tens of billions Many novel DDS that make use of these routes are beginning to enter
of pills are annually consumed worldwide for aspirin alone. Injections clinical trials and some have already reached the market. To accomplish
remain the primary mode of administering proteins and peptides. successful clinical translation, DDS must, at minimum, be safe, perform
More than 10 billion injections are performed each year worldwide their therapeutic function, offer convenient administration and offer
[1]. Oral pills offer convenience of pre-determined and measured ease of manufacturing. This review highlights some of the successful
doses, portability, defined dosing times and the overall non-invasive technologies that have made this transition (Fig. 1). Seven categories
nature of administration. However, they are also limited by the inability of DDS including microsphere-depots, tumor-targeting nanoparticles,
to deliver larger therapeutic molecules such as proteins [2]. Injections, transdermal patches, advanced oral pills, inhalers, implants and anti-
on the other hand, are able to deliver macromolecules, but are limited body–drug conjugates are highlighted. A search on clinicaltrials.gov
by their invasive nature and inapp.cropriate use [1]. Collectively, simple for: (i) ‘Depot’, (ii) ‘Transdermal’, (iii) ‘Inhaler’, (iv) ‘Subcutaneous
pills and injections are unable to meet many advanced therapeutic implant’ and ‘Intravitreal implant’ and ‘Birth control implant’, (v) ‘Nano-
needs including targeting, broad applicability to macromolecules and particle and cancer’, (vi) ‘Antibody drug conjugates’ and (vii) ‘OROS’®
on-demand activation. While not all pharmaceutical molecules require confirms high activity of clinical trials based on these categories of DDS
(Fig. 2). We discuss their historical perspective, advantages/limitations
in the clinic, the inspiration that they provide for follow-up technologies,
and current clinical status of new(er) products in the field. This review is
not intended to provide a comprehensive list of clinical and commercial
⁎ Corresponding author at: Department of Chemical Engineering, University of
status of all DDS given the high volume of activity in the field. Instead, the
California, Santa Barbara 93106, USA. Tel.: +1 805 893 7532; fax: +1 805 893 4731. article discusses select examples and analyzes their features that led to
E-mail address: samir@engineering.ucsb.edu (S. Mitragotri). their success.

http://dx.doi.org/10.1016/j.jconrel.2014.03.053
0168-3659/© 2014 Elsevier B.V. All rights reserved.
16 A.C. Anselmo, S. Mitragotri / Journal of Controlled Release 190 (2014) 15–28

2. Microparticle-based sustained release formulations thereby dramatically reducing the number of injections and increasing
both patient compliance and convenience. Reduced injection frequen-
Microparticle-based sustained release formulations have been cy leads to improved patient comfort and compliance, which are req-
developed to facilitate the controlled delivery of therapeutics. By uisites for successful self-administered DDS. In terms of the delivery
sustaining drug release over longer periods, these systems aim to im- technology, the individual components of Lupron Depot® offer several
prove the delivery of peptides or proteins by reducing injection fre- advantages. Specifically, PLGA polymer provides tunable degradation
quency [7]. Microparticle-based depots include a polymeric kinetics along with controlled release and well established safety
material (often biodegradable) that allows for protection of the and biocompatibility. Synthesis methods for Lupron Depot® micro-
drug cargo and control over drug release. A number of polymer particles must be highly reproducible and consistent in order to main-
choices exist, each with their own advantages and limitations. tain efficacy across patients. Indeed, the encapsulation of proteins and
Poly(lactic-co-glycolic) acid (PLGA), poly(lactic acid) (PLA) and peptides in PLGA particles has proven challenging in general, as is
polyglycolic acid (PGA) are perhaps the most commonly studied poly- maintaining protein stability in microparticles [12]. Lupron Depot®
mers due to their versatility in tuning biodegradation time and high bio- and, in general, all microparticle protein formulations face these
compatibility arising from their natural by-products, lactic acid and same challenges. Further, the production process of PLGA polymer de-
glycolic acid. Here, we will highlight one of the first Food and Drug Ad- termines product performance and different suppliers may not have
ministration (FDA)-approved, microparticle-based depot DDS, Lupron identical procedures which ties down drug companies to specific sup-
Depot®. plier(s). A variety of reasons, ranging from supplier shut down to the
high material costs, may pose a manufacturing challenge for
microparticles.
2.1. Development of Lupron Depot®

Lupron Depot® consists of leuprolide encapsulated in PLGA micro- 2.3. Lessons learned from Lupron Depot® — current academic research
spheres. Leuprolide was originally approved in 1985 as an injectable;
however, constant injections spurred interest in a more patient compli- As one of the first clinically and commercially successful peptide
ant formulation. It was long thought that controlled release of proteins, delivery microparticle depot DDS in the US, Lupron Depot® inspired
and even smaller peptides, from microspheres was impossible [8]. How- not only polymeric depot DDS, but also nanoparticle DDS in general.
ever, research in the mid-70s showed that this was indeed possible [9], Lupron Depot® is a perfect example of a polymeric controlled delivery
and thereby paved the way for a new class of peptide/protein encapsu- system that improves patient compliance by offering long-acting and
lated polymeric DDS. Lupron Depot® was one of the first examples of long-lasting alternatives to highly invasive (i.e. daily injections) thera-
this new class of controlled release polymeric DDS and was originally pies. Since the introduction of Lupron Depot®, researchers have ad-
developed by Takeda-Abbott Products, a joint venture formed in 1977 vanced the technology of sustained protein-release microparticles in
between Abbott Laboratories and Takeda, and approved by the FDA in various ways, including new methods for improving the stability and
1989 for the treatment of advanced prostate cancer [10]. Since then, protection of encapsulated proteins [13,14].
Lupron Depot® has been approved for management of endometriosis
and also for the treatment of central precocious puberty. Lupron
2.4. Current clinical landscape and future prospects
Depot® has been commercially successful, reaching annual sales of
nearly $1 billion [11].
Many other microparticle depot systems are in clinical use and have
been approved by the FDA (Table 2). One example is Nutropin Depot®
2.2. Advantages and limitations of Lupron Depot® developed by Genentech and Alkermes, the first long-acting formula-
tion for recombinant growth hormone. Nutropin Depot® is a biodegrad-
The main advantage of Lupron Depot® was that it significantly able microparticle depot formulation that was approved by the FDA in
lowered the number of injections required for the treatment. 1999 for pediatric growth hormone deficiency. Nutropin Depot® per-
Leuprolide alone required daily injections; however, the depot formu- formed well in preclinical studies, showing reliable delivery for over
lation requires injections every 1 to 6 months depending on the dose, one month in monkeys [15]. In clinical trials, Nutropin Depot® showed

Table 1
Routes of administration. Advantages, disadvantages, potential targets and examples of the most commonly used routes of administration for drug delivery. The number of top 100
commercial drugs and their route of administration were determined by counting the best selling drugs in 2013 as determined by Drugs.com [155].

Route of Advantages Disadvantages Targets Examples Number of top 100


administration commercial drugs

Injections: • Rapid onset (IV) • Difficult for patient to self-administer (IV) • Tissue with • Vaccines (IM) 42
IV, IM and SQ • Up to 100% bioavailability • Patients' fear of needles leads to noncompliance blood access (IV) • Chemotherapy
• Controlled depot release (IM, SQ) • Higher instance of infection • Systemic (IV)
• Suitable for most therapeutic molecules • Insulin (SQ)

Oral • Patient compliant and most convenient • Poor bioavailability • Systemic • Pills 54
• Generally nontargeted • Liquid
• Not viable for larger therapeutics (peptides/proteins) medications
• Potentially inconsistent due to the presence of food

Inhalation • Direct target to the lungs • Inconsistent delivery stemming from variation in • Lungs • Inhalers 7
• Fast absorption patient-to-patient technique • Brain • Anesthetics
• Systemic

Transdermal • Less side-effects due to direct delivery • Patients can potentially use incorrect dose (creams) • Skin • Patches 4
to the skin • Absorption dependent on skin condition and location • Systemic • Creams
• Bypasses first-pass degradation
A.C. Anselmo, S. Mitragotri / Journal of Controlled Release 190 (2014) 15–28 17

increase in the growth rate in children with pediatric growth hor-


mone deficiency while requiring only one to two doses a month,
compared to the standard of care which requires multiple injections
per week. Nutropin Depot® delivered a much larger molecule,
~ 22,000 Da, than the previously FDA approved microparticle depot
systems. Nutropin Depot®, however, is no longer commercially
available due to high cost and manufacturing challenges [16,17].
These issues are also encountered by other growth hormone prod-
ucts and, to some extent, several protein-based microparticle formula-
tions [18]. Despite improved patient compliance afforded by Nutropin
Depot®; this example highlights some of the after-market challenges
that face microparticle formulations that must be overcome in order to
remain competitive.
Alkermes has also developed Risperdal® Consta®, Vivitrol® and
Bydureon® which are FDA approved and clinically available micro-
sphere depot DDS. Risperdal® Consta®, approved in 2003 for the treat-
ment of schizophrenia, delivers Risperdal® for a two-week period. It
reduces inconsistencies associated with delivery using oral formula-
tions. Since its approval for schizophrenia, Risperdal® Consta® has re-
ceived approval for treatment of bipolar I disorder. Another Alkermes
product, Vivitrol®, which delivers naltrexone, was approved by the
FDA in 2006 for the treatment of alcohol dependence. Vivitrol® is
taken once a month as an alternative to the naltrexone oral formulation,
which suffered from severe compliance issues. As an anti-addiction
medication, Vivitrol® addresses compliance issues by providing a 30-
day treatment with a single injection. In 2010, Vivitrol® was approved
to treat opioid-dependent patients. Bydureon®, the most recently ap-
proved of these Alkermes products, was FDA approved in 2012 and de-
livers Exenatide for the treatment of type 2 diabetes. Bydureon® is a
weekly dose that provides an alternative to the twice daily injectable
form, Byetta®. Bydureon® has the advantage of long-lasting effects.
Other biodegradable, though non-microparticle, depot systems have
Fig. 1. Schematics and brief descriptions of the 7 highlighted DDS: (i) microparticle-based
been approved by the FDA. One example is Eligard®, an in situ forming
depot formulations, (ii) nanoparticle-based cancer drugs, (iii) transdermal systems
(patches highlighted here), (iv) oral drug delivery systems (OROS® highlighted here), biodegradable implant that delivers leuprolide for treatment of prostate
(v) pulmonary drug delivery systems (inhalers highlighted here), (vi) implants and cancer.
(vii) antibody–drug conjugates.
3. Nanoparticle-based cancer therapies

Nanoparticles (NPs) represent the most widely studied DDS. Over


the last 10 years, more than 25,000 publications (Web of Science key-
word: targeted nanoparticles, January 2014) have focused on targeted
drug delivery using NPs. Even within the field of NP-based drug delivery
systems, cancer remains the most studied target. NPs provide clear ad-
vantages for delivering chemotherapeutic drugs. NPs protect drugs
from degradation in vivo, provide targeting to diseased tissue, and con-
trol drug release at the target site. They also benefit from synthesis
methods that allow the scaled-up production of large batches of NPs
with near identical properties. The large number of publications in
this field is a combined result of the clinical need and the scientific dif-
ficulty that outlines the problem. The success rate, defined as the frac-
tion of new nanotherapies that lead to clinical products, however, is
very low. The path to FDA approval for nanomedicines is long and
risky; however, a handful of technologies have made strong strides
and are discussed here.

3.1. Doxil®: the first cancer nanomedicine

Doxil®, currently marketed by Johnson & Johnson, is the first FDA


approved liposomal NP formulation for the treatment of certain can-
cers [19] and has experienced commercial success, often exceeding
Fig. 2. Normalized pie chart for clinical trial search. Search on clinicaltrials.gov that counted hundreds of millions of dollars in sales per year [20]. Doxil® is a lipo-
the hits for clinical trials that are active and currently ongoing (but not recruiting). Thus, the somal formulation of doxorubicin and was approved by the FDA for
data present trials that are actually in process. Data has been normalized to the sum of the the treatment of AIDS-related Kaposi's sarcoma in 1995 and for re-
total hits (180) for the following search keywords: (i) ‘Depot’ (41), (ii) ‘Transdermal’ (38),
(iii) ‘Inhaler’ (27), (iv) ‘Subcutaneous implant’ (7) and ‘Intravitreal implant’ (9) and ‘Birth
current ovarian cancer in 1998 [21]. The development of Doxil®
control implant’ (10), (v) ‘Nanoparticle and cancer’ (25), (vi) ‘Antibody drug conjugates’ was enabled by research that shed light on various aspects of liposo-
(19) and (vii) ‘OROS’ (4). (Search conducted in Feb. 2014). mal formulations including long circulation [22], sufficient enhanced
18 A.C. Anselmo, S. Mitragotri / Journal of Controlled Release 190 (2014) 15–28

permeation and retention (EPR) effect [23], stable drug loading into performed well in initial clinical trials [37–39] and, in the years since,
liposomes [24] and releasing the encapsulated doxorubicin to target has been approved for treatment of various other cancers. Abraxane®
cells [25]. The early development efforts culminated in a study in has shown excellent commercial success with preliminary annual sales
Beagle dogs which confirmed that 2000 Da PEG–DSPE increased cir- in 2013 of $649 million [40].
culation time and reduced murine macrophage uptake in vitro better Other promising nanoparticle formulations have made the success-
than any of the other long-circulating alternatives [26]. These find- ful translation from academic labs to clinical trials. BIND Therapeutics
ings eventually led to initial “first in man” (FIM) studies for Doxil®. has developed a platform NP formulation (Accurins™) which combines
In these FIM studies, published in 1994, Doxil® exhibited a half-life active targeting ligands, protective and stealth functionality granted by
of about 45 h compared to about 10 h for free Doxorubicin. Further, PEG, controlled release via tuned polymer matrix and a therapeutic pay-
Doxil® showed marked enhancement (4- to 16-fold increase) of load into a single NP. Early studies with Accurins™ showed low toxicity
Doxorubicin delivery to tumors compared to free doxorubicin [27]. and stunted tumor growth in mice, comparable pharmacokinetics in
Doxil® was approved by the FDA in 1995 and was available clinically mice, rats and non-human primates and tumor shrinkage in some
in 1996. human patients in a clinical trial [41]. Accurins™ are currently in
Phase II clinical trials for treatment of non-small cell lung cancer and
3.2. Advantages and limitations of Doxil® prostate cancer. Another promising NP is CRLX101 (originally IT-101).
CRLX101 is a nanoparticulate chemotherapeutic formulation that con-
Doxil® offers many advantages over its free drug counterpart, doxo- sists of a camptothecin and cyclodextrin-based polymer conjugate.
rubicin. It exhibits enhanced circulation, increased tumor persistence CRLX101 benefits from the efficacy of camptothecin, the EPR targeting
and improved half-life compared to free doxorubicin [19]. Also, as a granted from the size of CRLX101 and sustained release of camptothecin.
nanoparticle, Doxil® can target certain tumors via the enhanced perme- These advantages limit many of the undesired side-effects of camp-
ability and retention (EPR) effect which allows small particles (~100 nm) tothecin while concomitantly providing a more stable formulation.
to accumulate in the leaky outer vasculature surrounding tumors [28]. The initial preclinical studies showed prolonged circulation time of
This increases preferential accumulation of NP around tumors and facil- CRLX101 compared to camptothecin alone, enhanced accumulation
itates NP delivery of drugs to tumors. This type of passive targeting sig- compared to camptothecin alone in tumors via the EPR effect and a
nificantly increases the amount of Doxil®-encapsulated doxorubicin prolonged release of camptothecin from CRLX101 following resi-
that reaches tumors compared to free doxorubicin. As a result, Doxil® dence in tumor tissue [42]. Early clinical studies point to promising
benefits from less off-target side effects and enhanced tumor killing. At safety, pharmacokinetic and efficacy results. CRLX101 is currently
the same time, there remain limitations. Despite being capable of EPR- being developed by Cerulean Pharma Inc. and is in Phase II clinical
mediated passive targeting, Doxil® lacks active targeting. The EPR effect trials [43]. Another cancer therapeutic that builds on the same plat-
is not exhibited to the same extent in all patients and can lead to differ- form technology of a drug conjugated to a cyclodextrin-based poly-
ences in the tumor targeting, and subsequent therapeutic efficacy. mer conjugate, CRLX301, is under development by Cerulean
Further, while Doxil® is PEGylated it is still cleared rapidly and could Pharma Inc. as well. In another example, Calando Pharmaceuticals
benefit from longer circulation, likely resulting in more passes around is developing a cyclodextrin-based NP formulation that is able to en-
the tumor and interactions with the target tissue. More recently, produc- capsulate and protect small-interfering RNA (siRNA) in sub-100 nm
tion issues of Doxil® have led to widespread shortage of Doxil® in the particles, CALAA-01. CALAA-01 is one of the first examples of an
US [29]. siRNA-based targeted nanoparticle used in clinical trials, and is com-
prised of: (i) linear cyclodextrin polymers that form the core, (ii)
3.3. Lessons learned from Doxil® — current academic research siRNA against RRM2 that inhibits growth of cancer cells, and surface
modification in the form of (iii) PEG coating, for stability, and (iv) a
As the first NP-related commercial success, Doxil® inspires current transferrin ligand, for targeting to cancer cells [44]. CALAA-01 is cur-
nanomedicine research, and in fact, many NP-based DDS attempt rently undergoing clinical trials and has shown promising results,
to improve upon Doxil®. Specifically, many NPs utilizing antibodies, specifically showing that: (i) CALAA-01 is able to localize in tumors,
peptides and various other ligands are being investigated to improve (ii) reduce the expression of RRM2 when compared to pre-CALAA-01
pharmacokinetics and target the delivery of therapeutics to specific dosed tissues and (iii) CALAA-01 mediates mRNA cleavage at pre-
sites in the body [30,31]. Concomitantly, stealth particles that avoid dicted sites [44]. These results collectively illustrate that siRNA sys-
rapid immune system clearance and circulate for longer times are temic administration, in nanoparticle form, can inhibit specific
being developed. Many strategies, ranging from hydrophilic coatings, genes in humans. Many other NP formulations that have made sig-
such as PEG, to macrophage-avoiding ligands such as CD47 are being nificant progress and are undergoing clinical trials have been exten-
used to functionalize NPs for stealth applications [32,33]. The mass pro- sively reviewed elsewhere [45–47]. Several in vitro transfection
duction of therapeutic NPs has benefited greatly from fabrication ad- reagents such as Lipofectamine 2000 have also been widely used in
vances in the semiconductor industry, as new methods utilizing mass in vitro cell cultures [48] and are not reviewed here.
producible nanofabrication techniques have been developed. A novel
technology, PRINT, makes it possible to mass produce specifically 4. Transdermal patches
sized, shaped, flexible, ligand-decorated and multi-layered NPs for ther-
apeutic application [34,35]. Transdermal patches offer a painless, patient-compliant interface to
facilitate the systemic administration of drugs. Many approved trans-
3.4. Current clinical landscape and future prospects dermal patches have a similar basic composition; a protective backing
layer that prevents drug leakage, a reservoir which stores the drug
Many nanoparticle-based therapies are being developed for the treat- which is ideally small and lipophilic, and an adhesive layer that facilitates
ment of cancer, yet few make it to a commercial product (Table 2). The skin contact [49]. Some systems may employ an extra layer that im-
most recent example is Marqibo®, liposomal vincristine, which was proves the controlled release of the drug. Many commercially successful
approved by the FDA in 2012 for treatment of a rare leukemia. Other transdermal patches have been developed for a variety of applica-
than Doxil®, perhaps the most well-known NP used in the clinic is tions ranging from birth control to smoking cessation. The first FDA-
Abraxane®, originally FDA approved in 2005 for the treatment of breast approved patch was a 3-day patch that delivered scopolamine for
cancer [36]. Abraxane®, currently marked by Celgene, is albumin-bound motion sickness treatment in 1979. Well over a dozen patches are cur-
paclitaxel, an already approved chemotherapy drug. Abraxane® rently in use and approved by the FDA (Table 2). Of the various patches,
Table 2
Examples of FDA approved drugs. List of approved drugs that fall into: (i) microparticle-based depot systems (biodegradable non-microparticle based formulations have been excluded), (ii) NP-based chemotherapies, (iii) transdermal devices [4],
(iv) osmotically controlled oral formulations [82], (v) inhalers [114], (vi) implants (emphasis on intravitreal, cancer, and birth control formulations) and (vii) antibody drug conjugates.

Microparticle-based NP-based Transdermal devices Osmotically controlled oral Inhalers Implants Antibody drug conjugates
depot chemotherapy formulations

Zmax® Abraxane® Transderm-Scop® (Scopolamine) Concerta® (Methylphenidate) Asthma and COPD: Eye: Kadcyla®
(Azithromycin) (Paclitaxel) Nitro-Dur® (Nitroglycerin) Ditropan XL® (Oxybutynin) Tudorza™ Pressair™ (Aclidinium) Vitrasert® (Trastuzumab emtansine)

A.C. Anselmo, S. Mitragotri / Journal of Controlled Release 190 (2014) 15–28


Decapeptyl®/ Doxil® Catapres-TTS® (Clonidine) Teczem (Enalapril Diltiazem) Proventil® HFA (Albuterol) (Ganciclovir) Adcetris®
Trelstar® (Doxorubicin) Estraderm® (Estradiol) Dilacor XR® (Diltiazem) Ventolin® HFA (Albuterol) Retisert® (Brentuximab vedotin)
(Triptorelin) DaunoXome® Duragesic® (Fentanyl) Covera-HS® (Verapamil) ProAir® HFA (Albuterol) (Fluocinolone)
Vivitrol® (Daunorubicin) Androderm® (Testosterone) DynaCirc CR® (Isradipine) Combivent Respimat® Ozurdex®
(Naltrexone) Marqibo® Combipatch® (Estradiol with norethindrone) Minipress® XL (Prazosin) (Albuterol and ipratropium) (Dexamethansone)
Arestin® (Vincristine) Lidoderm® (Lidocaine) Procardia XL® (Nifedipine) Fortamet® DuoNeb® (Albuterol and ipratropium)
(Minocycline) Climara Pro® (Estradiol with levonorgestrel) (Metformin) Altoprev® (Lovastatin) Brovana® (Arformoterol) Cancer:
Risperdal® Oxytrol® (Oxybutynin) Glucotrol XL® (Glipizide) QVAR® (Beclomethasone) Zoladex®
Consta® Synera® (Lidocaine and tetracaine) Invega® (Paliperidone) Pulmicort Flexhaler™ (Budesonide) (Goserelin)
(Risperidone) Daytrana® (Methylphenidate) Tegretol®-XL (Carbamazepine) Symbicort® (Budesonide and Formoterol) Gliadel®
Sandostatin® LAR Emsam® (Selegiline) Allegra D® (Pseudoephedrine and Alvesco® (Ciclesonide) (Prolifeprosan and
Depot (Octreotide) Neupro® (Rotigotine) Fexofenadine) Breo™ Ellipta™ (Fluticasone and vilanterol) Carmustine)
Nutropin Depot® Exelon® (Rivastigmine) Efidac/24® (Pseudoephedrine and Flovent® HFA (Fluticasone) Vantas®/Supprelin
(Somatropin) Sancuso® (Granisetron) Brompheniramine or Chlorphenir- Flovent® Diskus® (Fluticasone) LA® (Histrelin)
Lupron Depot® Butrans® (Buprenorphine) amine) Foradil® Aerolizer® (Formoterol) Viadur®
(Leuprolide) Ortho Evra® (Estradiol and norelgestromin) Volmax® (Albuterol) Perforomist® (Formoterol) (Leuprolide)
DepoCyt® Qutenza® (Capsaicin) Orenitram™ (Treprostinil) Arcapta™ Neohaler™ (Indacaterol)
(Cytarabine) Flector® (Diclofenac epolamine) Sudafed® 24 Hour (Pseudoephedrine) Atrovent® HFA (Ipratropium) Birth Control:
DepoDur® NicoDerm®/Habitrol®/ProStep™ (Nicotine) Exalgo® (Hydromorphone) Xopenex HFA™ (Levalbuterol) Nexplanon®
(Morphine) Asmanex® Twisthaler® (Mometasone) (Etonogestrel)
Bydureon® Dulera® (Mometasone and Formoterol) NuvaRing™
(Exenatide) Serevent™ Diskus™ (Salmeterol) (Etonogestrel and
Somatuline LA ADVAIR Diskus® (Salmeterol Fluticasone) ethinyl estradiol)
(Lanreotide) ADVAIR® HFA (Salmeterol Fluticasone) Mirena®/Norplant
Spiriva® Handihaler® (Tiotropium) (Levonorgestrel)
Paragard®
(Copper)
Devices: IONSYS® (Fentanyl) Other (PAH, cystic fibrosis):
SonoPrep® (Lidocaine via ultrasound) Cayston® (Aztreonam)
Iontocaine (Lidocaine and epinephrine via iontophoresis) Ventavis® (Iloprost)
LidoSite® (Lidocaine and epinephrine via iontophoresis) Tyvaso® (Treprostinil)
TOBI™ Podhaler ™ (Tobramycin)

19
20 A.C. Anselmo, S. Mitragotri / Journal of Controlled Release 190 (2014) 15–28

fentanyl stands out as an example of a commercially successful trans- skin surface and enhances transdermal transport. This technique
dermal patch used for chronic pain management. The fentanyl patch has been used to deliver several drugs including lidocaine [64], cy-
has exceeded $1 billion annual sales multiple times, highlighting its closporine A [65], hydrocortisone, dexamethasone and Ibuprofen
commercial success [50,51]. in humans [66].

4.1. Development of fentanyl patch 4.4. Current clinical landscape and future prospects

Fentanyl is a potent opioid analgesic first synthesized in 1960 [52] Newer, more sophisticated, transdermal patches and other trans-
and was quickly approved in Western Europe due to its rapid onset dermal DDS are being developed that are currently being tested in
and short duration, which made it an attractive choice as an analge- clinical trials. Notably, microneedles are investigated as a physical
sic for surgery. Alza Corporation and Janssen Pharmaceuticals began means of minimally piercing the skin, so as to avoid pain, prior to
exploration of a fentanyl transdermal patch to both increase the du- or jointly delivering a drug through the porated skin for systemic
ration of fentanyl and provide the patients with a potent opioid that delivery. Clinical studies have confirmed the ability of microneedles
could potentially be self-administered. A transdermal fentanyl patch, to deliver variety of drugs including insulin [67–69], lidocaine [70]
eventually known as Duragesic®, was developed by Alza Corpora- and vaccines [71]. Commercial development of microneedles is
tion and Janssen Pharmaceuticals and was approved by the FDA in also noteworthy. Zosano has developed a titanium microneedle
1990 for chronic pain [4]. This transdermal patch is a perfect exam- technology, applied via a reusable applicator, to deliver parathyroid
ple of a DDS that took an existing molecule (fentanyl) and improved hormone (PTH) across the skin [72] for the treatment of osteoporo-
its delivery, subsequently making it a commercial success. Several sis [73]. PTH delivery is desirable due to its potent bone building ef-
generic versions of fentanyl patches are also available. Another nota- fect, but it is currently only available as daily subcutaneous
ble commercially successful transdermal patch is Lidoderm®, which injections. Zosano Pharma-PTH (ZP-PTH) offers a much improved
was launched in 1999 and has reached annual sales of over $1 billion non-invasive delivery method and a longer shelf life compared to
in 2012 [53]. subcutaneous PTH injections. Phase II studies for ZP-PTH have
shown that ZP-PTH was effective in increasing spine bone mineral
4.2. Advantages and limitations of fentanyl patch density, in a dose dependent manner. Further, hip bone mineral
density was shown to increase at 6 months with ZP-PTH which
Duragesic® offered two main advantages over its counterpart, both has not been shown with the current approved subcutaneous treat-
of which improved patient compliance: (i) controlled release up to 72 ment [73]. ZP-PTH has finished both Phase I and Phase II trials [74].
h and (ii) formulated for self-administration. Fentanyl's early success Corium International Inc. is developing biodegradable microneedles
arose from the rapid onset time and short duration. By reformulating for PTH delivery.
fentanyl as a patch, the applications were broadened from surgery Other device-based technologies have also advanced from the lab to
room analgesic to potent, long lasting, pain relieving medication that the clinic (Table 2). For example, iontophoresis has been demonstrated
the patient could apply themselves as a predetermined dose. The lon- to deliver fentanyl and lidocaine in humans [75,76]. Commercial devices
ger acting, dose-optimized fentanyl patch reduced the need for multi- exist for topical delivery of lidocaine such as the active patch known as
ple doses and greatly improved patient compliance. Further, it offered LidoSite® developed by Vyteris. Clinical trials have shown that a ten
at-home and on-demand application without the need of an expert minute application delivered lidocaine via iontophoresis to induce
to administer the formulation. In the past, defects in the production effective anesthesia for venipuncture [76]. An ultrasound device,
of fentanyl patches have led to their recall and changes in the patch SonoPrep®, developed by Sontra Medical was also approved by the
design. FDA for delivery of lidocaine [77]. Systemic delivery of fentanyl using
iontophoresis has also made strong progress. IONSYS® was approved
4.3. Lessons learned from the fentanyl patch — current academic research by the FDA and the European Medicines Agency (EMA) in 2006. How-
ever, IONSYS® has not been launched in the US and was recalled in
The limitations of first generation transdermal patches have in- Europe. This technology is currently under development at Incline
spired the next generation of patches. The basic barrier function of therapeutics.
the skin inherently limits drug transport from transdermal patches. Methods are also being developed to use devices to perform contin-
The stratum corneum, the outermost layer of the skin, prevents the uous glucose monitoring across permeabilized skin. Symphony® (Echo
transport of molecules via its brick and mortar structure. Second Therapeutics) uses a non-invasive device to permeabilize the skin and
generation patches have been designed to perform the same func- then using a biosensor patch to monitor glucose levels for diabetic
tions as first generation patches but also enhance transport of the patients. It provides a means to avoid needle sticks and offer minute-
drug through the skin. A number of techniques, which can be incor- by-minute glucose monitoring with limited skin irritation in a compact
porated in or used alongside transdermal patches, are being devel- and lightweight package [78]. Symphony has demonstrated continuous
oped to enhance drug transport via transdermal patch. These transdermal glucose monitoring [79] and has applied for a CE mark.
include: (i) ultrasound [54], (ii) chemical permeation enhancers Microneedle-based technologies are also being developed for painless
[55], (iii) microneedles [56] and (iv) iontophoresis [57]. Chemical collection of blood. A device (Touch Activated Phlebotomy, TAP™) has
permeation enhancers disrupt the structure of the stratum corneum recently received a CE mark approval.
and significantly improve small molecule transport through this
barrier. Hundreds of permeation enhancers are known and have 5. Oral drug delivery
been studied for this application. Sophisticated methods for high
throughput screening of combinations of permeation enhancers Oral delivery is by far the most commonly used mode of drug admin-
have been developed and are currently used in industrial settings istration. This is evidenced by the number of oral formulations that
[58]. Iontophoresis uses electric fields to improve drug transport breached the top 100 sold drugs in 2013 (Table 1). Pre-determined
through the skin and the main advantage is the control over the doses, the simplicity offered by patient self-administration and systemic
quantity of drug transported, as it is directly proportional to the ap- delivery, all incorporated in one tablet or capsule, are the main advan-
plied current. This technique has been used to deliver opioids [59], tages of oral delivery. There are a variety of capsules in the market,
dopamine agonists [60], antiemetic agents [61], steroids [62] and and in fact, novel capsule designs offer an opportunity to develop
peptides [63] in humans. Ultrasound induces cavitation on the novel oral DDS. No peptides or proteins, however, are currently
A.C. Anselmo, S. Mitragotri / Journal of Controlled Release 190 (2014) 15–28 21

delivered by the oral route due to rapid degradation in the stomach and used specific enteric capsule coatings designed to degrade in specific
size-limited transport across the epithelium. Yet, much effort has sections of the GI tractor even co-administer enzyme inhibitors, both
been spent on developing oral delivery systems for proteins, especially with and without absorption enhancers, so as to avoid significant pro-
insulin. teolytic activity.

5.1. Development of OROS®-osmotically driven devices 5.4. Current clinical landscape and future prospects

Osmotically-driven DDS has been a topic of long-standing interest Oramed has developed a technology to deliver peptides and pro-
[80]. The interest in osmotically-driven pumps exhibited significant teins by oral administration. The technology makes use of protease in-
jump in the mid-70s [81]. The early designs of osmotic pumps eventually hibitors to prevent degradation of the protein in the stomach, an
led to the establishment of the osmotic-controlled release oral delivery absorption enhancer to facilitate passage through intestinal epithelium
system (OROS®) technology. The controlled delivery offered by OROS®, and an enteric coated capsule to ensure release of capsule contents in
which was developed by Alza, was tuned via a semi-permeable mem- the small intestine. Recent studies in humans showed that an Oramed
brane which surrounds the drug and allows for water penetration into insulin pill taken prior to each meal in type I diabetes patients, in con-
the drug reservoir. Water permeates into the drug reservoir and via junction with normal subcutaneous insulin injections, was able to re-
osmotic pumping, facilitates drug release from an open pore. The rate duce glycemia throughout the day [96]. Oramed has recently finished
of drug release is dependent on the osmotic properties of the drug and Phase II trials in the US. Emisphere is developing Eligen® technology
the design parameters can be tuned to control the release profile. The that can be applied for the oral delivery of molecules, including pro-
simplicity of the system paired with the advantages over simple oral teins. The Eligen® technology is based on compounds that interact
tablets led to a lasting impact of OROS® on oral drug delivery. The first with drugs and facilitate drug transport across the intestinal epithe-
product, Osmosin, was launched in Europe in 1982 and withdrawn in lium. Emisphere's technology has been used in clinical trials to
1983 due to severe GI tract irritation and, in some cases, intestinal wall transport insulin, salmon calcitonin, GLP-1 analogs and heparin via
perforation [82]. Since then, these issues have been addressed and, to the oral route [97]. Clinical trials have also shown that Eligen®-de-
date, numerous drugs in the clinic have been FDA approved and utilize livered oral vitamin B12 performs just as well as the standard B12
osmotically driven capsule systems (Table 2) with many more in clinical regimen of ~ 9 injections over a 90 day period [98]. Several other in-
trials. Collectively, these products speak for the high commercial impact novative technologies are being tested for oral delivery of proteins
of OROS®. Concerta®, one of approximately 10 products that utilize and are at various stages of development. These include the use of
OROS® technology, has generated over $1 billion in sales multiple mucoadhesive patches [99], FcRn-targeted nanoparticles [100] and
times [83]. hydrogels [101,102].

5.2. Advantages and limitations of OROS®


6. Pulmonary drug delivery
The main advantage of OROS® is that it allows controlled release of
therapeutics. Several simpler drug pills suffer from non-controlled Pulmonary drug delivery is mediated via the inhalation of drugs
drug release which leads to sharp peaks and valleys in pharmacokinet- through a variety of means, ranging from nebulizers to inhalers. Pulmo-
ic profiles. OROS® allowed improvements in terms of maintaining nary drug delivery has been explored for systemic as well as localized
drug concentration in plasma within a safe and therapeutic range delivery of drugs to the lungs. The advantages for systemic delivery
[84]. This allowed less frequent dosing and fewer side effects. Some are that inhalation offers a rapid systemic onset, in some cases on the
OROS® capsules are designed to release the drug in certain regions order of minutes, which results from the large surface area of lungs
of the GI tract which provides another layer of sophistication. As [103]. In the case of localized delivery to lungs, which has been the
with most oral DDS, OROS® offers a benefit of known predetermined primary application for inhaled medications, onset is even more rapid.
dose. Further, OROS® provides an avenue for the delivery of poorly sol- This is essential to treat severe asthma attacks as, in many cases, people
uble drugs, as evidenced by the success of Procardia XL® [82]. At the suffering from asthma attacks require simple to use/access (patient
same time, OROS® suffers from certain limitations that generally compliant) and fast acting DDS. Of the numerous inhalation DDS on
limit all oral delivery systems, namely poor delivery of peptides and the market, the metered dose inhalers have had the most impact, effec-
proteins. In addition, the localized drug release has been shown to tively making asthma medications simple for patients to administer to
cause GI irritation and ulcers [85], though such issues have been rela- themselves. The metered dose inhaler has also inspired other inhalation
tively rare [86]. DDS to push beyond what was originally thought possible, such as the
inhaled delivery of peptides or proteins. Here, we highlight the develop-
5.3. Lessons learned from OROS® — current academic research ment of the first metered dose inhaler.

Several oral drug delivery platforms have been developed, some in- 6.1. Development of asthma inhalers
spired by OROS®. These include protective coatings for the drug reser-
voir [87] and functionalized surfaces for mucoadhesion to intestinal The metered dose inhaler (MDI) was first conceived in the mid-50s
mucosa [88]. The primary excitement in the field of oral delivery, how- [104]. While other pulmonary DDS such as nebulizers had been devel-
ever, has been improving the ability to deliver proteins and peptides oped and used up to this point and found success due to localized
[89]. In general, most approaches have focused on improving protein lung delivery, they were limited as they were fragile and, most impor-
absorption in the GI tract or limiting proteolytic degradation of proteins tantly, not able to precisely control the amount of therapeutic given to
[2,90]. One method to improve these aspects is the use of mucoadhesive a patient. The MDI revolutionized pulmonary drug delivery by offering
systems that increase the residence time of oral DDS in the GI tract [91]. a controlled method to deliver a known amount of therapeutic with
Many mucoadhesive systems have been proposed and studied in small each dose. The first MDI was approved and marketed in 1956 [105].
animals, ranging from particle systems [92] to patches [93]. Other tech- The MDI design consists of a canister which contains the drug formula-
niques include using chemical permeation enhances to improve protein tion, the metering valve which dispenses the pre-determined dose and
absorption in the GI tract [94], targeting endocytotic pathways for a mouthpiece which facilitates the transport of the drug formulation to
enhanced cellular uptake [95] or enhancing the lipophilicity of proteins the patient. Nearly 10 years ago, it was estimated that asthma inhalers
to improve partitioning into epithelial cell membranes [2]. Some have generate sales of about $25 billion a year [106]. More recently, the top
22 A.C. Anselmo, S. Mitragotri / Journal of Controlled Release 190 (2014) 15–28

two selling combination inhalers, Advair® and Symbicort®, generated The first FDA approved inhalable insulin formulation was Nektar's
$8 and $3 billion, respectively, in 2012 [83]. Exubera®, which was manufactured and marketed by Pfizer. The
Exubera® formulation stored and delivered insulin via a dry powder
6.2. Advantages and limitations of inhalers inhaler [115] so as to maintain peptide stability. Further, Exubera® pro-
vided appropriate aerosolized characteristics to ensure pulmonary de-
MDIs offer numerous advantages for delivery of asthma medication livery, a low-dose powder filling so as to deliver small doses and a
over nebulizers. The inability of older nebulizers to control the amount physical device to control dispersion and reliable dosing to patients
of therapeutic dose resulted in increased, potentially fatal, side effects of [116]. Despite successful clinical trials [117,118], the additional cost
overdose or insufficient delivery. These inconsistencies allowed MDI in- of Exubera® [119] caused the discontinuation of the product [120].
halers to provide much needed ease of administration in terms of both MannKind Inc. continued to develop an inhaled insulin product,
consistent dosing and simple application procedures. The combination AFREZZA® which potentially offers a compact inhalation device, low
of the large dispersion area of aerosolized DDS that typically follows dosing and rapid absorption [121]. AFREZZA® has submitted a New
administration and the large surface area of the lungs synergistically Drug Application (NDA) and has recently announced positive Phase III
contribute to the delivery of inhaled therapeutics by facilitating the clinical study results, namely weight advantages, reduced hypoglycemia
rapid uptake of molecules into systemic circulation. Further, if local and decrease in fasting blood glucose levels compared to insulin aspart
delivery to lungs is desired, inhalers provide a direct access. However, [122].
inhalers are limited in their ability to deliver peptides and proteins. Deg- Other future prospects for inhalable DDS focus on taking advantage
radation of stored peptides and proteins is a particular issue for MDIs. of either: (i) the rapid drug onset for near immediate treatment or
Dry powder inhalers (DPIs) are potentially more advantageous than (ii) the localized delivery to lungs for the treatment of lung infections.
MDIs for protein storage as they allow for the storage of proteins in a Civitas is developing a pulmonary delivery platform called ARCUS®
dry form, effectively extending protein shelf life. Further, DPIs are typi- which is focused on treating OFF episodes associated with Parkinson's
cally activated by the patient's air flow [107] as compared to MDIs disease with a formulation known as CVT-301. CVT-301 is designed to
which requires coordination between actuation and inhalation. The deliver L-dopa and is to be used as needed and in tandem with stan-
patient is responsible for this coordination and must know the proper dard L-dopa oral formulated Parkinson's disease treatments. The
ways to administer their own medication; however, this issue can be ARCUS® inhaler is designed to provide immediate relief from OFF epi-
addressed with proper training. sodes. Briefly, ARCUS® provides reliable delivery of a large, precise
dose via a breath activated device. ARCUS® utilizes a dry powder for-
mulation that ensures both stability and delivery deep into the lungs
6.3. Lessons learned from inhalers — current academic research for systemic delivery. Phase I studies have shown that CVT-301 was suc-
cessful in achieving target L-dopa plasma levels and Phase II studies
Recent research has improved the use of MDIs and other inhalers by: have shown improved motor functions in Parkinson's disease patients
(i) optimizing aerosol particle size for ideal aerodynamic diameters for that were in the OFF state [123]. Currently, CVT-301 is undergoing
enhanced local delivery to lungs [108], (ii) engineering of spacers to Phase II studies to determine safety and efficacy in treatment of OFF
limit the loss of drug at target site during application [104] and (iii) im- states in Parkinson's disease patients. Cardeas is developing inhalable
proving on the actual design and technology of the original MDI, to antibiotics for pneumonia treatment using a single-use nebulizer to de-
name a few [109]. In a broader sense, inhalation research is focused on liver two synergistically acting antibiotics, amikacin and fosfomycin.
the delivery of larger molecules like peptides via the inhalation route Cardeas was recently granted fast track review for their antibiotic com-
for systemic administration. Inhalation is an attractive route for protein bination product and has begun enrollment for Phase II studies for
delivery due to the rapid absorption of drugs through the lungs, the lack treatment of multi-drug resistant bacterial infections [124]. Early clini-
of proteolytic enzymes in lungs compared to GI tract and the general cal trials indicate that the Cardeas formulation is safe and tolerable for
non-invasive nature of inhalers. Techniques have been developed to all tested doses.
improve macromolecule delivery via inhalation including absorption
enhancers [110] and improved particle design for either deep lung pen-
etration [111] or extended delivery of encapsulated therapeutics [112]. 7. Implantable systems

6.4. Current clinical landscape and future prospects Implantable DDS can be classified into two categories that define
their release properties; either passive delivery or active delivery.
Inhalers have been predominately used, and FDA approved, for Passive DDS control drug release via the material properties that
the treatment of asthma and chronic obstructive pulmonary disease constitute the implant. Passive DDS can tune drug release from the
(COPD) (Table 2). As of December 2013, the FDA has completed the reservoir by controlling rates of diffusion, osmosis, or concentration
phase-out of chlorofluorocarbon inhalers [113], with many products gradients. These passive release methods are dependent on drug choice,
still remaining [114]. Many other devices have been approved or are membrane composition, size and tortuosity of membrane pores, and
in development for the treatment of other diseases including cystic the combination of these design parameters. On the other hand, active
fibrosis (Cayston® and TOBI™ Podhaler™), diabetes (Exubera®, implant DDS control drug release using a pump that can be activated
approved in 2006 and withdrawn in 2007) and pulmonary arterial hy- by a number of methods ranging from simple manual actuation from
pertension (Ventavis® and Tyvaso®) (Table 2) [115]. Inhalation offers physical pressure to electrochemically driven mechanisms that can
a broad platform that can systemically deliver peptides and proteins vary drug delivery rates. Since some implantable DDS take advantage
in a patient-compliant, non-invasive manner. In particular, they offer of micro and nano-fabrication technologies, this may lead to longer
rapid absorption, lesser interference from proteases compared to oral approval times as they may fall under the FDA Office of Combination
route and high permeability of the epithelium. These advantages over Product review, which looks to review medical therapies that
oral route for peptide and protein delivery influence the landscape of combines elements from independently established fields [125].
clinical inhalation research, as the search for non-invasive delivery of Implantable DDS have made a large impact in ocular drug delivery
peptides and proteins continues to persist. Many inhalable DDS, as as traditional methods require numerous intravitreal injections which
highlighted below, are making strong strides to achieve routine clinical can potentially result in a number of issues as discussed below. Here,
delivery of insulin. In fact, one such product had been previously ap- we will highlight the very first of these intravitreal implants, the
proved by the FDA. Vitrasert® implant.
A.C. Anselmo, S. Mitragotri / Journal of Controlled Release 190 (2014) 15–28 23

7.1. Development of Vitrasert® can be fabricated via semiconductor technology for more precise and
reliable synthesis.
The Vitrasert® implant was approved by the FDA in 1996 for the
treatment of AIDS-related cytomegalovirus (CMV) retinitis. The con- 7.4. Current clinical landscape and future prospects
ventional means of delivering ganciclovir has been intravitreal injec-
tions; however, the nature of CMV retinitis treatment requires many Other implantable DDS have been developed and approved by the
injections that result in complications that can arise from repeated in- FDA (Table 2), and a few examples are highlighted here. Iluvien®, a
travitreal injections. Theoretically, reservoir implants provide an attrac- microfabrication-based device has been developed to deliver fluocinolone
tive means to deliver ganciclovir while limiting the number of acetonide. Iluvien® does not require suturing as in the case with both
intravitreal procedures, but no such device existed at the time. The Vitrasert® and Retisert®, which provides a much less invasive procedure.
Vitrasert® implant consists of a ganciclovir reservoir that is coated in The implant is designed to last up to 3 years without requiring replace-
both polyvinyl alcohol (PVA) and ethylene vinyl acetate (EVA) that is ment. Iluvien® delivers fluocinolone acetonide to patients suffering
sutured on the inside wall of the eye. The PVA mediates the release of from diabetic macular edema, which is typically treated via laser photoco-
drug and the EVA controls the surface area that the drug can pass agulation that often leaves patients with irreversible blind spots. Similar
through. The implant can maintain controlled drug release for a period to its predecessors, Iluvien® targets a disease that offers virtually no alter-
of 5–8 months, after which the implant must be removed and replaced native in the form of FDA approved drug therapies. Iluvien® has been
in order to continue treatment. In early studies, Vitrasert® showed zero approved and is being marketed in many countries including the UK,
order release of ganciclovir [126] and in a Phase I trial showed efficacy Germany and Spain and is currently being considered for FDA approval.
for controlling CMV retinitis in patients that showed little to no re- Ongoing Phase II clinical trials aimed at treating wet-age related macular
sponse to conventional ganciclovir therapies [127]. Vitrasert® was edema and dry-age related macular edema are also under way. Another
eventually advanced by Control Delivery Systems and has been ap- FDA approved implant, Ozurdex®, is used to deliver corticosteroid dexa-
proved by the FDA. methasone for treatment of retinal vein occlusion. Ozurdex® is the first,
and only, biodegradable implant that delivers dexamethasone [134]. Fol-
lowing drug release from the implant, no surgery is required to remove
7.2. Advantages and limitations of Vitrasert®
the implant from the body.
The Gliadel® wafer was FDA approved in 1996 for use as an ad-
Vitrasert® was the first implant approved for CMV retinitis treat-
junct to surgery in patients with recurrent glioblastoma multiforme,
ment. The alternative, intravitreal ganciclovir injections, require fre-
an aggressive type of brain tumor. In 2003, the Gliadel® wafer
quent (weekly) administration over a period of months or years [128].
was approved for use as a first time treatment for brain tumors. It
The issues associated with these repeated intravitreal injections ranged
was the first localized chemotherapeutic for initial treatment of
from hemorrhaging to retinal detachment [129]. Further, rapid drug
brain cancer. The dime sized wafers, which are comprised of the che-
clearance from the vitreous region led to more frequent injections, on
motherapeutic agent carmustine and the polymer matrix made of
the order of 1–2 per week [128]. The Vitrasert® implant circumvents
poly(carboxyphenoxy-propane/sebacic acid) [135], are surgically
the majority of these issues. Controlled release allows for better drug
inserted in the cavity that remains following removal of brain tumors.
absorption which lowers the number of required procedures compared
Gliadel® has shown to increase patient survival for up to 6 months
to intravitreal injections. This results in improved sustained release and
in some cases, but also increased incidences of postoperative wound
better intraocular concentrations over a longer period of time compared
infections and cerebral edema [136]. Current clinical Gliadel® re-
to both combination of initial intravitreal injection alongside mainte-
search looks to combine Gliadel® wafers with systemic chemother-
nance intravenous injections [128]. Other advantages include localized
apies and radiation treatment for enhanced tumor cell death
delivery to the eye and specified dosing. Vitrasert® lasts between 5
following surgery.
and 8 months before replacement is necessary. The initial clinical trials
MicroCHIPS is developing a platform of implantable DDS, which uti-
showed that ganciclovir implants limited progression of CMV retinitis
lizes a MEMS-based technology for drug delivery [137]. These implants
nearly 3-fold longer than intravenous ganciclovir alone could offer
allow active control over drug release, either at predetermined times or
[130]. The disadvantages of Vitrasert® include the lack of systemic
on-demand. Release of PTH has been demonstrated using MicroCHIPs
treatment as CMV infection can also appear as a systemic infection
technology and offers precise dosing at scheduled intervals to overcome
and oral supplementation of ganciclovir is often required to prevent
the compliance issues associated with the daily injections conventional
CMV extraocular infection. Two separate invasive surgeries for the im-
PTH treatments currently require. Human studies have shown that PTH
plantation and removal of Vitrasert® are required and, independent of
was delivered as programmed and showed similar pharmacokinetics
the Vitrasert® implant, can cause complications due to the nature of
while offering less variation in dosing as compared to multiple PTH
eye surgery.
injections. Further, bone formation was increased in the short, 20 day,
study [138].
7.3. Lessons learned from Vitrasert® — current academic research
8. Antibody–drug conjugates
Retisert®, approved by the FDA in 2005 for the treatment of chron-
ic non-infectious uveitis, followed Vitrasert® as the second generation Antibody–drug conjugates (ADCs) are targeted bioconjugate
of reservoir based implants. Retisert® provides reliable drug delivery pharmaceuticals that combine the benefits of monoclonal antibodies
for up to 3 years, well past that of the 5–8 month limit of Vitrasert®. and cytotoxic drugs to treat cancer. ADCs involve a chemical linker
Retisert® is also smaller compared to Vitrasert® which makes implan- that conjugates the antibody to the drug and can be either cleavable
tation and removal less invasive. Both of these products address areas or non-cleavable. Indeed, the vast number of monoclonal antibodies
of the pharmaceutical market where there is truly no competitive al- and drugs allow for near endless combinations of ADCs. ADCs poten-
ternative that offers long-term drug release. More recently, active re- tially offer a development path for highly toxic drugs that are other-
lease implants which offer on demand drug release have been wise difficult to clinically implement due to off-target toxicity. As
developed in academic labs [131–133]. These devices can control an example, the ADC Brentuximab vedotin (Adcetris®) facilitated
drug release via a variety of mechanisms including, but not limited the use of the highly toxic monomethyl auristatin E (MMAE) in the
to: physical forces, electrochemical methods, magnetic fields, laser in- clinic, which is otherwise challenging due to the high toxicity of
duced or electrothermal means. Further, currently researched devices MMAE. To date, three ADCs have been approved by the FDA; the
24 A.C. Anselmo, S. Mitragotri / Journal of Controlled Release 190 (2014) 15–28

first one, Gemtuzumab ozogamicin (Mylotarg®), has been with- encoding unnatural amino acids into antibodies for site-specified drug
drawn from the market in 2010. Here, we will highlight the develop- binding [145]. Optimization of ADCs has been investigated for over
ment of the most recently approved ADC, Trastuzumab emtansine 20 years [146] and so the majority of research efforts are focused on
(commercialized by Roche), which has seen early commercial ADCs currently in clinical trials [147,148].
success.
8.4. Current clinical landscape and future prospects
8.1. Development of Trastuzumab emtansine (Kadcyla®)
There are over 25 clinical trials currently underway for ADCs [147,
Trastuzumab emtansine (Kadcyla®) is comprised of the monoclonal 148]. Many of them utilize the same, or similar, cytotoxic drugs as
antibody trastuzumab, the potent cytotoxic drug mertansine (DM1) the two currently approved ADCs, Kadcyla® and Adcetris®. The other
and a linker that covalently attaches trastuzumab and DM1 together currently FDA approved ADC, Adcetris®, was approved in 2011 for
via reactive succinimide ester and maleimide groups. Trastuzumab is a the treatment of Hodgkin's lymphoma and anaplastic large cell lym-
monoclonal antibody (FDA approved in 1998) that is used for the treat- phomas. Adcetris® is comprised of a CD30-specific antibody, a highly
ment of certain breast cancers by targeting HER2 receptors. The main potent antitubulin agent monomethyl auristatin E (MMAE) and an en-
limitation of trastuzumab is eventual tumor resistance. Combination zyme cleavable linker that releases the drug when inside the target
of trastuzumab with DM1 can address this issue. DM1, is a derivative cells [149]. The combination of these provides a potent therapy, as
of maytansine which are known to be a toxic class of therapeutics and the drug component of Adcetris®, MMAE, is too potent to have
have essentially no therapeutic window [139]. The derivative itself, any clinical utility on its own and the antibody portion, cAC10, has lim-
DM1, is 100 to 10,000 fold more potent than typical chemotherapeutics ited therapeutic effect on its own. Adcetris® generated sales of
[139], which limits clinical use of DM1 to targeted applications. The $136 million in its first year on the market (October 2011 to September
linker component utilizes activated maleimide reactive groups on 2012) [150].
trastuzumab's surface to bind to thiol groups on DM1 [140]. The Gemtuzimab ozogamicin (Mylotarg®) was the first FDA approved
Phase III studies of Kadcyla® showed an increase in progression- ADC; however, it was withdrawn from the market in 2010, 10 years
free survival of 3.2 months, overall survival of 5.8 months and after its initial approval. The market withdrawal was spurred by a clin-
lower toxicity when compared to treatment with lapatinib plus cap- ical trial that indicated that Mylotarg® not only increased mortality, but
ecitabine in patients that were previously treated with trastuzumab also added no benefit over alternative therapies [151]. Mylotarg® was
and a taxane [141]. Kadcyla® was approved by the FDA in 2013 and used for the treatment of acute myelogenous leukemia and consisted
generated a revenue of over $150 million within a few months of of an antibody directed toward CD33, a highly potent cytotoxic
launch [142]. calicheamicin derivative and an acid-labile hydrazone linker [152].
Mylotarg® is still being tested in clinical trials and showing promise in
8.2. Advantages and limitations of Trastuzumab emtansine combination with chemotherapy [153]. As stated earlier, over 25 ADCs
are currently in clinical trials [148], and some of the ones in later devel-
Kadcyla® offers the benefits of a highly potent cytotoxic drug opment stages include: (i) Inotuzumab ozogamicin, developed by
alongside the targeted abilities of the monoclonal antibody Pfizer, which targets CD22 with a calicheamicin payload attached via a
trastuzumab. The main advantages are being able to combine two hydrazone linker, (ii) Glembatumumab vedotin which targets trans-
separately established technologies to synergistically increase the membrane protein NMB with an MMAE agent attached via an enzyme
therapeutic efficacy beyond what is offered by individual compo- cleavable dipeptide and has received FDA Fast Track designation for
nents. The ADC directly addresses the clinical limitations of each breast cancer treatment and (iii) Lorvotuzumab mertansine which tar-
individual composition, namely the lack of long term efficacy of anti- gets CD56 with DM1 attached via a cleavable disulfide linker and has
bodies upon repeated dosing and the lack of clinical utility afforded been granted orphan drug status for the treatment of small cell lung
the highly potent DM1 drug due to its high toxicity which typically cancer.
results from lack of specific tumor targeting. What remains is a highly
potent targeted therapy, which chemotherapy cannot provide, for the 9. Summary and outlook
treatment of HER2 positive breast cancer. ADCs offer a larger therapeu-
tic window and lower side effects compared to traditional chemother- There is no doubt that DDS have had a strong impact on pharma-
apies, stemming from their selective tumor targeting. Unfortunately, ceutical market. Novel DDS have provided several advantages in-
the main issues that challenge successful monoclonal antibodies also cluding local delivery, use of drugs that are otherwise difficult to
limit ADCs, specifically: (i) poor access to hypoxic tumor areas and use and increased patient compliance. In addition, drug delivery sys-
generally poor tumor penetration, (ii) issues concerning the non- tems also provide means of patent extension, which further adds the
target site uptake, and (iii) undesirable immune system responses via value of new drug molecules. A review of the field reveals several
Fc interactions [143]. clinical and commercial success stories. Many sub-fields of drug de-
livery, i.e., transdermal drug delivery, depot injections, and oral drug
8.3. Lessons learned from Kadcyla® — current academic research delivery, have commercially successful products with annual sales
exceeding a billion dollars. At the same time, it should also be
The early success of Kadcyla® highlights the importance of selecting noted that the journey from an idea to a commercially successful
a highly specific antibody that had previously shown clinical success DDS is long and difficult, as evidenced by the few FDA approved
and the selection of a highly toxic drug that likely could not be delivered products in spite of a strong early research pipeline. At each step
in its free, non-targeted, form. Research continues to focus on the linker there are many roadblocks that can spell immediate failure (Fig. 3),
technology that conjugates the drug to the antibody. This is because thus preventing the clinical or commercial translation of many promis-
both highly specific and highly toxic drugs already exist, so cutting ing DDS.
edge research is focused on linker technology that can directly improve Strong academic research has provided numerous early-stage tech-
ADCs by improving homogenous attachment between antibody and nologies for DDS. Naturally, many commercially successful DDS began
drug, thereby facilitating more predictable circulation, cell internaliza- as novel ideas originating in an academic research lab. The hypotheses
tion and drug release of ADCs. Some specific efforts involve chemically that form the foundation of these ideas need to be tested and verified
limiting the number of potential binding sites on antibodies to have as soon as possible. Proof of concept studies are essential in confirming
better control of drug–antibody stoichiometry [144] and genetically that the DDS is viable and can provide therapeutic benefits in vivo,
A.C. Anselmo, S. Mitragotri / Journal of Controlled Release 190 (2014) 15–28 25

substantial capital and require outside sources of funding and sup-


port in order to progress the technology. Further, companies likely
have access to therapeutic molecules that are in need of novel drug
delivery systems. Clinical trials are the ultimate test of efficacy and
utility of a novel DDS. Prior to FDA approval, DDS candidates must
show efficacy and tolerability in pre-clinical studies. Even after suc-
cessful studies and commercial launch, side-effects still need to be
monitored and recorded, as it is difficult to cover all details in clinical
trials. This is evidenced by the recall of many drugs and DDS formu-
lations after commercialization. Indeed, many DDS fail when under-
going clinical trials for a variety of reasons, ranging from: (i) lack of
funds to support expensive clinical trials, (ii) lack of tolerability or
high toxicity in patients even in spite of therapeutically successful
outcomes, (iii) lack of therapeutic effect or (iv) study design failures
Fig. 3. The typical path that academic discoveries follow from initial ideation to a final stemming from patient selection, dosing issues or even selecting an
commercialized product. The challenges at each step are highlighted. appropriate endpoint. Timeline of development also needs to be
closely monitored in view of the finite patent life and extended reg-
ulatory approval times.
Many of the DDS showcased here (Table 3) were developed or
inspired by research at the academic level. While the process from
even if just in small animals. Given the high rate of fall-out during lab-level research to a commercial product is long, there are many
in vitro to in vivo translation, it is critical that the idea be put to the examples that illustrate the potential of ideas and proof-of-concept
‘in vivo test’ as soon as possible. In vitro optimization of the DDS studies in building the foundation for these commercialized products
must typically occur in order to provide the best chance for success or even facilitating the development of new sub-fields in drug deliv-
at expensive, and often lengthy, in vivo experiments. Further, success ery. Partnering between pharmaceutical companies and academic
in small animal models does not guarantee success in humans. Fol- labs has the potential to further both fundamental drug delivery re-
lowing up with publications and patents is also key. Strong publica- search and translate these findings in the form of improved clinical
tions often provide validation of significant scientific breakthroughs therapeutics.
through peer-review [154] while patents are essential to secure
commercial development.
Investors and industrial partners play a strong role in commercial Acknowledgments
as well as clinical translation. While the two do not necessarily
have to correlate, the high costs of clinical studies and often re- This work was supported by the National Institutes of Health under
quires strong financial commitments. Unlike technologies originat- grant no. R01DK097379 and National Science Foundation Graduate
ed in large pharmaceutical companies, academic inventions lack Research Fellowship under grant no. DGE-1144085 to ACA.

Table 3
Case study summary. Advantages, disadvantages and specific contributions to drug delivery of the 7 highlighted DDS.

DDS Case study Advantages compared General disadvantages Case study contribution
(target disease) to alternatives

Microparticle depots Lupron Depot® • Longer-lasting • Potential production issues • One of the first examples of controlled peptide delivery
(cancer) • Lowers injection frequency • Changed the way peptide/proteins can be delivered

Nanoparticles for cancer Doxil® (cancer) • EPR targeted • Lacks active targeting • First FDA approved intravenous cancer nanoparticle
treatment • Long-circulating • EPR effect can be unpredictable
• Less side-effects

Transdermal devices Duragesic® • Extended release • Limited permeability through • Took a relevant therapeutic and provided a means for
(pain relief) • Patient Compliant the skin widespread self-administration
• Designated Dose

Oral delivery systems OROS® (various, • Designated Dose • Limited delivery of peptides • Platform technology that is still relevant and delivers
see Table 2) • Patient Compliant and proteins new therapeutics over 30 years later
• Unidirectional release led to
GI irritation and ulcers

Pulmonary delivery MDI inhalers • Designated Dose • Requires patient trained in • Transformed pulmonary delivery by offering controlled,
systems (asthma) • Patient Compliant inhaler use on-demand delivery
• Challenging to delivery large
molecules

Implants Vitrasert • Lasts 5–8 months instead of weekly • Invasive surgery for insertion • Limited the number of invasive procedures in favor or
(CMV retinitis) • Limits number of injections/ and removal less-invasive, longer-lasting implants
procedures done to the eye • Potentially lacks systemic
treatment

Antibody drug conjugates Kadcyla® • Both targeted and highly potent • New DDS and not yet optimized • DDS that utilized an already FDA approved molecule
(cancer) • ADCs can use already FDA approved • Same issues that face mAb (Trastuzumab) to permit the delivery of highly
molecules toxic agents
26 A.C. Anselmo, S. Mitragotri / Journal of Controlled Release 190 (2014) 15–28

References [38] M. Green, et al., Abraxane®, a novel Cremophor®-free, albumin-bound particle


form of paclitaxel for the treatment of advanced non-small-cell lung cancer, Ann.
[1] M. Kermode, Unsafe injections in low-income country health settings: need for Oncol. 17 (8) (2006) 1263–1268.
injection safety promotion to prevent the spread of blood-borne viruses, Health [39] D.W. Nyman, et al., Phase I and pharmacokinetics trial of ABI-007, a novel nanopar-
Promot. Int. 19 (1) (2004) 95–103. ticle formulation of paclitaxel in patients with advanced nonhematologic malig-
[2] M. Goldberg, I. Gomez-Orellana, Challenges for the oral delivery of macromole- nancies, J. Clin. Oncol. 23 (31) (2005) 7785–7793.
cules, Nat. Rev. Drug Discov. 2 (4) (2003) 289–295. [40] Celgene Corporation Announces 2014 Financial Outlook and Preliminary 2013
[3] J.S. Patton, P.R. Byron, Inhaling medicines: delivering drugs to the body through the Results, (February 9, 2014) Available from: http://ir.celgene.com/releasedetail.
lungs, Nat. Rev. Drug Discov. 6 (1) (2007) 67–74. cfm?ReleaseID=821044.
[4] M.R. Prausnitz, R. Langer, Transdermal drug delivery, Nat. Biotechnol. 26 (11) [41] J. Hrkach, et al., Preclinical development and clinical translation of a PSMA-targeted
(2008) 1261–1268. docetaxel nanoparticle with a differentiated pharmacological profile, Sci. Transl.
[5] R. Gaudana, et al., Ocular drug delivery, AAPS J. 12 (3) (2010) 348–360. Med. 4 (128) (2012) 128ra39.
[6] L. Illum, Nasal drug delivery: new developments and strategies, Drug Discov. Today [42] S. Svenson, et al., Preclinical to clinical development of the novel camptothecin
7 (23) (2002) 1184–1189. nanopharmaceutical CRLX101, J. Control. Release 153 (1) (2011) 49–55.
[7] J.E. Talmadge, The pharmaceutics and delivery of therapeutic polypeptides and [43] S. Eliasof, et al., Correlating preclinical animal studies and human clinical trials of a
proteins, Adv. Drug Deliv. Rev. 10 (2) (1993) 247–299. multifunctional, polymeric nanoparticle, Proc. Natl. Acad. Sci. U. S. A. 110 (37)
[8] R. Langer, Controlled release of a therapeutic protein, Nat. Med. 2 (7) (1996) 742–743. (2013) 15127–15132.
[9] R. Langer, J. Folkman, Polymers for the sustained release of proteins and other [44] M.E. Davis, et al., Evidence of RNAi in humans from systemically administered
macromolecules, Nature 263 (5580) (1976) 797–800. siRNA via targeted nanoparticles, Nature 464 (7291) (2010) 1067–1070.
[10] J.C. Wright, A.S. Hoffman, Historical overview of long acting injections and [45] A.Z. Wang, R. Langer, O.C. Farokhzad, Nanoparticle delivery of cancer drugs, Annu.
implants, Long Acting Injections and Implants, Springer, 2012, pp. 11–24. Rev. Med. 63 (2012) 185–198.
[11] M. Chaubal, Polylactides/glycolides—excipients for injectable drug delivery and [46] Y. Matsumura, K. Kataoka, Preclinical and clinical studies of anticancer agent-
beyond, Drug Deliv. Technol. 2 (2002) 34–36. incorporating polymer micelles, Cancer Sci. 100 (4) (2009) 572–579.
[12] G. Zhu, S.R. Mallery, S.P. Schwendeman, Stabilization of proteins encapsulated in [47] J.D. Heidel, M.E. Davis, Clinical developments in nanotechnology for cancer thera-
injectable poly (lactide-co-glycolide), Nat. Biotechnol. 18 (1) (2000) 52–57. py, Pharm. Res. 28 (2) (2011) 187–199.
[13] V. Sinha, A. Trehan, Biodegradable microspheres for protein delivery, J. Control. [48] B. Dalby, et al., Advanced transfection with Lipofectamine 2000 reagent: primary
Release 90 (3) (2003) 261–280. neurons, siRNA, and high-throughput applications, Methods 33 (2) (2004) 95–103.
[14] M. van de Weert, W.E. Hennink, W. Jiskoot, Protein instability in poly (lactic-co- [49] S. Venkatraman, R. Gale, Skin adhesives and skin adhesion. 1. Transdermal drug
glycolic acid) microparticles, Pharm. Res. 17 (10) (2000) 1159–1167. delivery systems, Biomaterials 19 (13) (1998) 1119–1136.
[15] O.L. Johnson, et al., A month-long effect from a single injection of microencapsulated [50] A.C. Watkinson, A commentary on transdermal drug delivery systems in clinical
human growth hormone, Nat. Med. 2 (7) (1996) 795–799. trials, J. Pharm. Sci. 102 (9) (2013) 3082–3088.
[16] W. Jiang, et al., Biodegradable poly(lactic-co-glycolic acid) microparticles for inject- [51] T.H. Stanley, Fentanyl, J. Pain Symptom Manage. 29 (5 Suppl) (2005) S67–S71.
able delivery of vaccine antigens, Adv. Drug Deliv. Rev. 57 (3) (2005) 391–410. [52] T.H. Stanley, The history and development of the fentanyl series, J. Pain Symptom
[17] F. Jordan, et al., Sustained release hGH microsphere formulation produced by a Manage. 7 (3 Suppl) (1992) S3–S7.
novel supercritical fluid technology: in vivo studies, J. Control. Release 141 (2) [53] Lidoderm Sales Data, (February 9 2014) Available from: http://www.drugs.com/
(2010) 153–160. stats/lidoderm.
[18] M.A. Tracy, Development and scale-up of a microsphere protein delivery system, [54] S. Mitragotri, Healing sound: the use of ultrasound in drug delivery and other ther-
Biotechnol. Prog. 14 (1) (1998) 108–115. apeutic applications, Nat. Rev. Drug Discov. 4 (3) (2005) 255–260.
[19] Y. Barenholz, Doxil(R)—the first FDA-approved nano-drug: lessons learned, J. Control. [55] P. Karande, A. Jain, S. Mitragotri, Insights into synergistic interactions in binary
Release 160 (2) (2012) 117–134. mixtures of chemical permeation enhancers for transdermal drug delivery,
[20] Johnson & Johnson Annual Report 2012, (February 9 2014). Available from: J. Control. Release 115 (1) (2006) 85–93.
https://www.jnj.com/sites/default/files/pdf/JNJ2012annualreport.pdf. [56] M.R. Prausnitz, Microneedles for transdermal drug delivery, Adv. Drug Deliv. Rev.
[21] T.M. Allen, P.R. Cullis, Drug delivery systems: entering the mainstream, Science 303 56 (5) (2004) 581–587.
(5665) (2004) 1818–1822. [57] Y.N. Kalia, et al., Iontophoretic drug delivery, Adv. Drug Deliv. Rev. 56 (5) (2004)
[22] T. Allen, A. Chonn, Large unilamellar liposomes with low uptake into the reticulo- 619–658.
endothelial system, FEBS Lett. 223 (1) (1987) 42–46. [58] P. Karande, A. Jain, S. Mitragotri, Discovery of transdermal penetration enhancers
[23] Y. Matsumura, H. Maeda, A new concept for macromolecular therapeutics by high-throughput screening, Nat. Biotechnol. 22 (2) (2004) 192–197.
in cancer chemotherapy: mechanism of tumoritropic accumulation of [59] M.A. Ashburn, et al., Iontophoretic delivery of morphine for postoperative analgesia,
proteins and the antitumor agent smancs, Cancer Res. 46 (12 Part 1) (1986) J. Pain Symptom Manage. 7 (1) (1992) 27–33.
6387–6392. [60] R. van der Geest, et al., Iontophoretic delivery of apomorphine. II: An in vivo study
[24] G. Haran, et al., Transmembrane ammonium sulfate gradients in liposomes in patients with Parkinson's disease, Pharm. Res. 14 (12) (1997) 1804–1810.
produce efficient and stable entrapment of amphipathic weak bases, Biochim. [61] M. Cormier, et al., Effect of transdermal iontophoresis codelivery of hydrocortisone
Biophys. Acta (BBA)—Biomembr. 1151 (2) (1993) 201–215. on metoclopramide pharmacokinetics and skin-induced reactions in human
[25] A.T. Horowitz, Y. Barenholz, A.A. Gabizon, In vitro cytotoxicity of liposome- subjects, J. Pharm. Sci. 88 (10) (1999) 1030–1035.
encapsulated doxorubicin: dependence on liposome composition and [62] M.P. James, R.M. Graham, J. English, Percutaneous iontophoresis of prednisolone—a
drug release, Biochim. Biophys. Acta (BBA)—Biomembr. 1109 (2) (1992) pharmacokinetic study, Clin. Exp. Dermatol. 11 (1) (1986) 54–61.
203–209. [63] P.G. Green, Iontophoretic delivery of peptide drugs, J. Control. Release 41 (1)
[26] A.A. Gabizon, Y. Barenholz, M. Bialer, Prolongation of the circulation time of doxo- (1996) 33–48.
rubicin encapsulated in liposomes containing a polyethylene glycol-derivatized [64] E.L. Spierings, J.A. Brevard, N.P. Katz, Two‐minute skin anesthesia through ultra-
phospholipid: pharmacokinetic studies in rodents and dogs, Pharm. Res. 10 (5) sound pretreatment and iontophoretic delivery of a topical anesthetic: a feasibility
(1993) 703–708. study, Pain. Med. 9 (1) (2008) 55–59.
[27] A. Gabizon, et al., Prolonged circulation time and enhanced accumulation in malig- [65] P. SantoiannI, M. Nino, G. Calabro, Intradermal Drug Delivery by Low Frequency
nant exudates of doxorubicin encapsulated in polyethylene-glycol coated lipo- Sonophoresis (25 kHz), 2004.
somes, Cancer Res. 54 (4) (1994) 987–992. [66] B.E. Polat, et al., Ultrasound-mediated transdermal drug delivery: mechanisms,
[28] H. Maeda, et al., Tumor vascular permeability and the EPR effect in macromolecular scope, and emerging trends, J. Control. Release 152 (3) (2011) 330–348.
therapeutics: a review, J. Control. Release 65 (1) (2000) 271–284. [67] J.J. Norman, et al., Faster pharmacokinetics and increased patient acceptance of
[29] Important Update Regarding Supply Outage of DOXIL®, Available from: http:// intradermal insulin delivery using a single hollow microneedle in children and
www.doxil.com/doxil-supply-shortage February 9 2014. adolescents with type 1 diabetes, Pediatr. Diabetes 14 (6) (2013) 459–465.
[30] D. Peer, et al., Nanocarriers as an emerging platform for cancer therapy, Nat. [68] J. Gupta, E.I. Felner, M.R. Prausnitz, Rapid pharmacokinetics of intradermal insulin
Nanotechnol. 2 (12) (2007) 751–760. administered using microneedles in type 1 diabetes subjects, Diabetes Technol.
[31] E. Ruoslahti, Peptides as targeting elements and tissue penetration devices for Ther. 13 (4) (2011) 451–456.
nanoparticles, Adv. Mater. 24 (28) (2012) 3747–3756. [69] J. Gupta, E.I. Felner, M.R. Prausnitz, Minimally invasive insulin delivery in subjects
[32] P.L. Rodriguez, et al., Minimal “self” peptides that inhibit phagocytic clearance and with type 1 diabetes using hollow microneedles, Diabetes Technol. Ther. 11 (6)
enhance delivery of nanoparticles, Science 339 (6122) (2013) 971–975. (2009) 329–337.
[33] S.M. Moghimi, A.C. Hunter, J.C. Murray, Long-circulating and target-specific [70] J. Gupta, et al., Rapid local anesthesia in humans using minimally invasive
nanoparticles: theory to practice, Pharmacol. Rev. 53 (2) (2001) 283–318. microneedles, Clin. J. Pain 28 (2) (2012) 129–135.
[34] J.P. Rolland, et al., Direct fabrication and harvesting of monodisperse, shape- [71] Y.C. Kim, J.H. Park, M.R. Prausnitz, Microneedles for drug and vaccine delivery, Adv.
specific nanobiomaterials, J. Am. Chem. Soc. 127 (28) (2005) 10096–10100. Drug Deliv. Rev. 64 (14) (2012) 1547–1568.
[35] S.W. Morton, et al., Scalable manufacture of built-to-order nanomedicine: spray- [72] M. Ameri, S.C. Fan, Y.F. Maa, Parathyroid hormone PTH(1–34) formulation that
assisted layer-by-layer functionalization of PRINT nanoparticles, Adv. Mater. 25 enables uniform coating on a novel transdermal microprojection delivery system,
(34) (2013) 4707–4713. Pharm. Res. 27 (2) (2010) 303–313.
[36] M. Ferrari, Cancer nanotechnology: opportunities and challenges, Nat. Rev. Cancer [73] P.E. Daddona, et al., Parathyroid hormone (1–34)-coated microneedle patch
5 (3) (2005) 161–171. system: clinical pharmacokinetics and pharmacodynamics for treatment of osteo-
[37] W.J. Gradishar, et al., Phase III trial of nanoparticle albumin-bound paclitaxel com- porosis, Pharm. Res. 28 (1) (2011) 159–165.
pared with polyethylated castor oil-based paclitaxel in women with breast cancer, [74] A. Arora, M.R. Prausnitz, S. Mitragotri, Micro-scale devices for transdermal drug
J. Clin. Oncol. 23 (31) (2005) 7794–7803. delivery, Int. J. Pharm. 364 (2) (2008) 227–236.
A.C. Anselmo, S. Mitragotri / Journal of Controlled Release 190 (2014) 15–28 27

[75] S. Mayes, M. Ferrone, Fentanyl HCl patient-controlled iontophoretic transdermal [112] J.S. Patton, J. Bukar, S. Nagarajan, Inhaled insulin, Adv. Drug Deliv. Rev. 35 (2–3)
system for the management of acute postoperative pain, Ann. Pharmacother. 40 (1999) 235–247.
(12) (2006) 2178–2186. [113] FDA to Complete Phase-out of Chlorofluorocarbon Inhalers, (February 9, 2014,
[76] W.T. Zempsky, et al., Evaluation of a low-dose lidocaine iontophoresis system for Published: October 23, 2013) Available from: http://www.fda.gov/newsevents/
topical anesthesia in adults and children: a randomized, controlled trial, Clin. newsroom/pressannouncements/ucm371901.htm.
Ther. 26 (7) (2004) 1110–1119. [114] Drug Treatments for Asthma and Chronic Obstructive Pulmonary Disease That
[77] B.M. Becker, et al., Ultrasound with topical anesthetic rapidly decreases pain of Do Not Use Chlorofluorocarbons, (February 9, 2014) Available from: http://
intravenous cannulation, Acad. Emerg. Med. 12 (4) (2005) 289–295. www.fda.gov/Drugs/DrugSafety/InformationbyDrugClass/ucm082370.htm.
[78] S.K. Vashist, Non-invasive glucose monitoring technology in diabetes management: [115] A.J. Hickey, Back to the future: inhaled drug products, J. Pharm. Sci. 102 (4) (2013)
a review, Anal. Chim. Acta. 750 (2012) 16–27. 1165–1172.
[79] H. Chuang, et al., Pilot studies of transdermal continuous glucose measurement [116] S. White, et al., EXUBERA: pharmaceutical development of a novel product for
in outpatient diabetic patients and in patients during and after cardiac surgery, pulmonary delivery of insulin, Diabetes Technol. Ther. 7 (6) (2005) 896–906.
J. Diabetes Sci. Technol. 2 (4) (2008) 595–602. [117] T. Quattrin, et al., Efficacy and safety of inhaled insulin (Exubera) compared with
[80] S. Rose, J.F. Nelson, A continuous long-term injector, Aust. J. Exp. Biol. Med. Sci. 33 subcutaneous insulin therapy in patients with type 1 diabetes: results of a 6-
(4) (1955) 415–419. month, randomized, comparative trial, Diabetes Care 27 (11) (2004) 2622–2627.
[81] F. Theeuwes, Elementary osmotic pump, J. Pharm. Sci. 64 (12) (1975) 1987–1991. [118] P.A. Hollander, et al., Efficacy and safety of inhaled insulin (exubera) compared with
[82] V. Malaterre, et al., Oral osmotically driven systems: 30 years of development and subcutaneous insulin therapy in patients with type 2 diabetes: results of a 6-month,
clinical use, Eur. J. Pharm. Biopharm. 73 (3) (2009) 311–323. randomized, comparative trial, Diabetes Care 27 (10) (2004) 2356–2362.
[83] E. Rosenthal, The Soaring Cost of a Simple Breath, (February 9, 2014), Published: [119] C. Black, et al., The clinical effectiveness and cost-effectiveness of inhaled insulin in
October 12, 2013]; Available from: http://www.nytimes.com/2013/10/13/us/the- diabetes mellitus: a systematic review and economic evaluation, Health Technol.
soaring-cost-of-a-simple-breath.html?_r=0. Assess. 11 (33) (2007) 1–126.
[84] F. Theeuwes, Orososmotic system development, Drug Dev. Ind. Pharm. 9 (7) [120] A. Opar, Another blow for inhaled protein therapeutics, Nat. Rev. Drug Discov. 7 (3)
(1983) 1331–1357. (2008) 189–190.
[85] C.G. Park, et al., A nanofibrous sheet-based system for linear delivery of nifedipine, [121] J. Kling, Dreamboat sinks prospects for fast approval of inhaled insulin, Nat.
J. Control. Release 149 (3) (2011) 250–257. Biotechnol. 29 (3) (2011) 175–176.
[86] D.M. Bass, M. Prevo, D.S. Waxman, Gastrointestinal safety of an extended-release, [122] Press Releases: MannKind Reports Positive Data from a Phase 3 Clinical Study of
nondeformable, oral dosage form (OROS): a retrospective study, Drug Saf. 25 AFREZZA in Patients with Type 1 Diabetes, (February 9, 2014, Published: August
(14) (2002) 1021–1033. 14, 2013) Available from: http://www.news.mannkindcorp.com/phoenix.zhtml?
[87] G.A. Agyilirah, G.S. Banker, Polymers for Enteric Coating Applications. Polymers for c=147953&p=irol-newsArticle&ID=1847441&highlight=.
Controlled Drug Delivery, CRC Press, Boca Raton, 1991. 39–66. [123] Available from: http://www.civitastherapeutics.com/CVT-301.html.
[88] G. Ponchel, J.-M. Irache, Specific and non-specific bioadhesive particulate systems [124] Cardeas Pharma Announces that FDA Grants QIDP Designation and Fast Track
for oral delivery to the gastrointestinal tract, Adv. Drug Deliv. Rev. 34 (2) (1998) Review for the Company's Antibiotic Combination Product, (February 9, 2014,
191–219. Published: January 8, 2014) Available from: http://cardeaspharma.com/
[89] M. Morishita, N.A. Peppas, Is the oral route possible for peptide and protein drug press-releases/cardeas-pharma-announces-that-fda-grants-qidp-designation-
delivery? Drug Discov. Today 11 (19) (2006) 905–910. and-fast-track-review-for-the-companys-antibiotic-combination-product/.
[90] A. Bernkop-Schnürch, The use of inhibitory agents to overcome the enzymatic bar- [125] M. Staples, et al., Application of micro- and nano-electromechanical devices to
rier to perorally administered therapeutic peptides and proteins, J. Control. Release drug delivery, Pharm. Res. 23 (5) (2006) 847–863.
52 (1) (1998) 1–16. [126] T.J. Smith, et al., Intravitreal sustained-release ganciclovir, Arch. Ophthalmol. 110
[91] L.M. Ensign, R. Cone, J. Hanes, Oral drug delivery with polymeric nanoparticles: the (2) (1992) 255–258.
gastrointestinal mucus barriers, Adv. Drug Deliv. Rev. 64 (6) (2012) 557–570. [127] G.E. Sanborn, et al., Sustained-release ganciclovir therapy for treatment of cyto-
[92] E. Mathiowitz, et al., Biologically erodable microspheres as potential oral drug megalovirus retinitis. Use of an intravitreal device, Arch. Ophthalmol. 110 (2)
delivery systems, Nature 386 (6623) (1997) 410–414. (1992) 188–195.
[93] V. Gupta, et al., Mucoadhesive intestinal devices for oral delivery of salmon calcitonin, [128] G. Velez, S.M. Whitcup, New developments in sustained release drug delivery
J. Control. Release 172 (3) (2013) 753–762. for the treatment of intraocular disease, Br. J. Ophthalmol. 83 (11) (1999)
[94] P.D. Ward, T.K. Tippin, D.R. Thakker, Enhancing paracellular permeability by 1225–1229.
modulating epithelial tight junctions, Pharm. Sci. Technol. Today 3 (10) (2000) [129] D.H. Geroski, H.F. Edelhauser, Drug delivery for posterior segment eye disease,
346–358. Invest. Ophthalmol. Vis. Sci. 41 (5) (2000) 961–964.
[95] P.W. Swaan, Recent advances in intestinal macromolecular drug delivery via [130] D.C. Musch, et al., Treatment of cytomegalovirus retinitis with a sustained-release
receptor-mediated transport pathways, Pharm. Res. 15 (6) (1998) 826–834. ganciclovir implant. The Ganciclovir Implant Study Group, N. Engl. J. Med. 337 (2)
[96] R. Eldor, et al., Glucose-reducing effect of the ORMD-0801 oral insulin preparation (1997) 83–90.
in patients with uncontrolled type 1 diabetes: a pilot study, PLoS One 8 (4) (2013) [131] C.L. Stevenson, J.T. Santini Jr., R. Langer, Reservoir-based drug delivery systems
e59524. utilizing microtechnology, Adv. Drug Deliv. Rev. 64 (14) (2012) 1590–1602.
[97] The Eligen® Library, (February 9, 2014) Available from: http://www.emisphere.com/ [132] R.A. Receveur, F.W. Lindemans, N.F. de Rooij, Microsystem technologies for
eligen_library.html. implantable applications, J. Micromech. Microeng. 17 (5) (2007) R50.
[98] M.C. Castelli, et al., Comparing the efficacy and tolerability of a new daily oral vita- [133] A.C. Richards Grayson, et al., Electronic MEMS for triggered delivery, Adv. Drug
min B12 formulation and intermittent intramuscular vitamin B12 in normalizing Deliv. Rev. 56 (2) (2004) 173–184.
low cobalamin levels: a randomized, open-label, parallel-group study, Clin. Ther. [134] How Ozurdex(R) Works, (March 11, 2014) Available from: http://www.ozurdex.com/
33 (3) (2011) 358–371 (e2). HowItWorks.aspx.
[99] K. Whitehead, Z. Shen, S. Mitragotri, Oral delivery of macromolecules using intestinal [135] A. Giese, et al., Pattern of recurrence following local chemotherapy with biodegrad-
patches: applications for insulin delivery, J. Control. Release 98 (1) (2004) 37–45. able carmustine (BCNU) implants in patients with glioblastoma, J. Neurooncol. 66
[100] E.M. Pridgen, et al., Transepithelial transport of fc-targeted nanoparticles by the (3) (2004) 351–360.
neonatal fc receptor for oral delivery, Sci. Transl. Med. 5 (213) (2013) 213ra167. [136] E.L. Weber, E.A. Goebel, Cerebral edema associated with Gliadel wafers: two case
[101] M. Torres‐Lugo, et al., pH‐sensitive hydrogels as gastrointestinal tract absorption studies, Neuro Oncol. 7 (1) (2005) 84–89.
enhancers: transport mechanisms of salmon calcitonin and other model molecules [137] J.T. Santini Jr., M.J. Cima, R. Langer, A controlled-release microchip, Nature 397
using the Caco‐2 cell model, Biotechnol. Prog. 18 (3) (2002) 612–616. (6717) (1999) 335–338.
[102] M. Torres-Lugo, et al., Physicochemical behavior and cytotoxic effects of [138] R. Farra, et al., First-in-human testing of a wirelessly controlled drug delivery
p (methacrylic acid–g-ethylene glycol) nanospheres for oral delivery of proteins, microchip, Sci. Transl. Med. 4 (122) (2012) 122ra21.
J. Control. Release 80 (1) (2002) 197–205. [139] B. Hughes, Antibody–drug conjugates for cancer: poised to deliver? Nat. Rev. Drug
[103] R.U. Agu, et al., The lung as a route for systemic delivery of therapeutic proteins and Discov. 9 (9) (2010) 665–667.
peptides, Respir. Res. 2 (4) (2001) 198. [140] J.R. Junutula, et al., Site-specific conjugation of a cytotoxic drug to an antibody
[104] R. Dalby, J. Suman, Inhalation therapy: technological milestones in asthma treat- improves the therapeutic index, Nat. Biotechnol. 26 (8) (2008) 925–932.
ment, Adv. Drug Deliv. Rev. 55 (7) (2003) 779–791. [141] S. Verma, et al., Trastuzumab emtansine for HER2-positive advanced breast cancer,
[105] P.J. Anderson, History of aerosol therapy: liquid nebulization to MDIs to DPIs, N. Engl. J. Med. 367 (19) (2012) 1783–1791.
Respir. Care 50 (9) (2005) 1139–1150. [142] ImmunoGen, Inc., Reports First Quarter Fiscal Year 2014 Financial Results and
[106] J.B. Fink, B.K. Rubin, Problems with inhaler use: a call for improved clinician and Provides Quarterly Update, (February 9, 2014, Published: October 25, 2013) Avail-
patient education, Respir. Care 50 (10) (2005) 1360–1375. able from: http://investor.immunogen.com/releasedetail.cfm?ReleaseID=800370.
[107] M.J. Telko, A.J. Hickey, Dry powder inhaler formulation, Respir. Care 50 (9) (2005) [143] P. Chames, et al., Therapeutic antibodies: successes, limitations and hopes for the
1209–1227. future, Br. J. Pharmacol. 157 (2) (2009) 220–233.
[108] D.A. Edwards, A. Ben-Jebria, R. Langer, Recent advances in pulmonary drug delivery [144] C.F. McDonagh, et al., Engineered antibody–drug conjugates with defined sites and
using large, porous inhaled particles, J. Appl. Physiol. 85 (2) (1998) 379–385. stoichiometries of drug attachment, Protein Eng. Des. Sel. 19 (7) (2006) 299–307.
[109] M.B. Dolovich, R. Dhand, Aerosol drug delivery: developments in device design and [145] J.Y. Axup, et al., Synthesis of site-specific antibody–drug conjugates using unnatu-
clinical use, Lancet 377 (9770) (2011) 1032–1045. ral amino acids, Proc. Natl. Acad. Sci. U. S. A. 109 (40) (2012) 16101–16106.
[110] A. Hussain, et al., Absorption enhancers in pulmonary protein delivery, J. Control. [146] S.C. Alley, N.M. Okeley, P.D. Senter, Antibody–drug conjugates: targeted drug delivery
Release 94 (1) (2004) 15–24. for cancer, Curr. Opin. Chem. Biol. 14 (4) (2010) 529–537.
[111] J.L. Cleland, A. Daugherty, R. Mrsny, Emerging protein delivery methods, Curr. [147] J.M. Lambert, Drug-conjugated antibodies for the treatment of cancer, Br. J. Clin.
Opin. Biotechnol. 12 (2) (2001) 212–219. Pharmacol. 76 (2) (2013) 248–262.
28 A.C. Anselmo, S. Mitragotri / Journal of Controlled Release 190 (2014) 15–28

[148] A. Mullard, Maturing antibody–drug conjugate pipeline hits 30, Nat. Rev. Drug [153] A.K. Burnett, et al., The addition of gemtuzumab ozogamicin to intensive chemo-
Discov. 12 (5) (2013) 329–332. therapy in older patients with AML produces a significant improvement in overall
[149] A. Younes, et al., Brentuximab vedotin (SGN-35) for relapsed CD30-positive lym- survival: results of the UK NCRI AML16 randomized trial, Blood, 2011, (AMER SOC
phomas, N. Engl. J. Med. 363 (19) (2010) 1812–1821. HEMATOLOGY 1900 M STREET. NW SUITE 200, WASHINGTON, DC 20036 USA).
[150] R.S. Zolot, S. Basu, R.P. Million, Antibody–drug conjugates, Nat. Rev. Drug Discov. 12 [154] R. Langer, A personal account of translating discoveries in an academic lab, Nat.
(4) (2013) 259–260. Biotechnol. 31 (6) (2013) 487–489.
[151] FDA: Pfizer Voluntarily Withdraws Cancer Treatment Mylotarg from U.S. Market, [155] U.S. Pharmaceutical Sales — 2013, (January 17, 2014) Available from: http://www.
(February 9, 2014 Published: June 21, 2010) Available from: http://www.fda.gov/ drugs.com/stats/top100/2013/sales.
NewsEvents/Newsroom/PressAnnouncements/ucm216448.htm.
[152] A.M. Wu, P.D. Senter, Arming antibodies: prospects and challenges for
immunoconjugates, Nat. Biotechnol. 23 (9) (2005) 1137–1146.

You might also like