throbber
Received: 23 January 2016
`
`DOI 10.1002/btm2.10003
`
`| Accepted: 25 February 2016
`
`R E V I E W
`
`Nanoparticles in the clinic
`
`Aaron C. Anselmo1
`
`| Samir Mitragotri2
`
`1David H. Koch Institute for Integrative
`Cancer Research, Massachusetts Institute of
`Technology, Cambridge, MA 02139
`
`2Dept. of Chemical Engineering, Center for
`Bioengineering, University of California,
`Santa Barbara, CA 93106
`
`Correspondence
`Samir Mitragotri, Dept. of Chemical
`Engineering, University of California, Santa
`Barbara, CA 93106.
`Email: samir@engineering.ucsb.edu
`
`Funding information
`SM acknowledges support from the
`National Institute of Health
`(1R01HL129179-01).
`
`Abstract
`Nanoparticle/microparticle-based drug delivery systems for systemic (i.e., intravenous) applications
`have significant advantages over their nonformulated and free drug counterparts. For example,
`nanoparticle systems are capable of delivering therapeutics and treating areas of the body that
`other delivery systems cannot reach. As such, nanoparticle drug delivery and imaging systems are
`one of the most investigated systems in preclinical and clinical settings. Here, we will highlight the
`diversity of nanoparticle types, the key advantages these systems have over their free drug coun-
`terparts, and discuss their overall potential in influencing clinical care. In particular, we will focus
`on current clinical trials for nanoparticle formulations that have yet to be clinically approved. Addi-
`tional emphasis will be on clinically approved nanoparticle systems, both for their currently
`approved indications and their use in active clinical trials. Finally, we will discuss many of the often
`overlooked biological, technological, and study design challenges that impact the clinical success of
`nanoparticle delivery systems.
`
`K E Y W O R D S
`clinic, translational medicine, clinical translation, clinical trials, drug delivery, nanomedicine,
`nanoparticles
`
`1 |
`
`I N T R O D U C T I O N
`
`Nanoparticle/microparticle delivery systems are widely investigated pre-
`clinically with many particle-based formulations and technologies having
`already been introduced in the clinic.1–5 Oral, local, topical, and systemic
`(e.g., intravenous) administration are all proven methods that have been
`Food and Drug Administration (FDA)-approved for the delivery of nano-
`particles/microparticles, depending on the desired application or tar-
`geted site. For example: (a) oral delivery of particles has been approved
`clinically for imaging applications (e.g., Gastromark),6 (b) local delivery of
`particles has been widely used in the clinic as depot delivery systems for
`the extended delivery of a variety of biologics including peptides and
`other small molecules (e.g., DepoCyt),4 (c) topical application of particles
`has been approved clinically to increase penetration of biologics across
`the skin barrier (e.g., Estrasorb),7 and (d) systemic delivery of particles
`has been approved clinically for treating a variety of cancers (e.g., Doxil)8
`and other diseases. Given the utility and success of these clinical exam-
`ples, preclinical research efforts for each of these delivery methods con-
`tinue to increase with particular attention placed on developing new
`applications and further improving their delivery and efficacy.
`
`Of these delivery methods, intravenously administered nanoparticles
`receive the most attention, both preclinically and clinically. The increased
`interest for intravenous delivery is not surprising given that nanoparticles
`delivered systemically have direct access to nearly all parts of the body and
`thus have the most potential to influence clinical care. For this same reason,
`systemically delivered nanoparticles also face exceedingly difficult chal-
`lenges with regards to both the delivery aspect (e.g., biological challenges)9,10
`and the regulatory aspect (e.g., study design and approval challenges).11,12
`This review focuses on the clinical translation of intravenously administered
`nanoparticles, with additional emphasis on the challenges faced by nanopar-
`ticles from a clinical and translational point of view. Specifically, the biologi-
`cal, technological, and study design challenges facing the clinical translation
`of nanoparticles will be discussed. Comprehensive lists of intravenous nano-
`particle technologies that are either approved or currently in clinical trials
`will be provided to highlight the current clinical landscape.
`
`2 | N A N O P A R T I C L E T Y P E S , A P P L I C A T I O N S ,
`A D VA N T A G E S , A N D P O T E N T I AL
`
`Therapeutic and diagnostic nanoparticles typically fall into two catego-
`ries: (a) inorganic nanoparticles (e.g., gold, silica, iron oxide, etc.) and (b)
`
`VC 2016 The Authors. Bioengineering & Translational Medicine is published by Wiley Periodicals, Inc. on behalf of The American Institute of Chemical Engineers. This is an
`open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium, provided the original
`work is properly cited.
`
`10 |
`
`wileyonlinelibrary.com/btm2
`
`Bioengineering & Translational Medicine 2016; 1: 10-29
`
`

`

`ANSELMO AND MITRAGOTRI
`
`| 11
`
`F I G U R E 1 Clinically relevant nanoparticles. Organic and inorganic nanoparticles have been approved for a variety of clinical indications (black text)
`and are being investigated in current clinical studies for additional indications (red text). Examples included (a) Doxil (200 nm scale bar), (b) Abraxane
`(200 nm scale bar), (c) CRLX101 (50 nm scale bar), (d) Feraheme (20 nm scale bar), (e) early iteration of Cornell Dots (50 nm scale bar), and (f) gold
`nanoshells (inset: 100 nm scale bar, main figure: 1,000 nm scale bar) from Nanospectra, makers of AuroLase. (a) Reprinted from ref. 16. Copyright
`(2016), with permission from Elsevier. (b) Adapted by permission from Macmillan Publishers Ltd: Nature Communications,17 copyright (2015). (c)
`Reprinted from ref. 18 (d) Reprinted from refs. 16 and 19. Copyright (2016), with permission from Elsevier. (e) Adapted with permissions from ref.
`20. Copyright (2012) American Chemical Society. (f) Reprinted from ref. 21
`
`organic nanoparticles (e.g., polymeric, liposomes, micelles, etc.). Inor-
`ganic nanoparticles have been successful
`in preclinical studies, are
`being developed in the clinic for a variety of applications including
`intraoperative sentinel lymph node imaging and thermal ablation of
`tumors, and have already been approved for imaging applications and
`anemia treatment (Figure 1).13–15 Alongside this, organic nanoparticles
`have also exhibited substantial success in the clinic where they are
`currently being developed for broad applications ranging from vaccina-
`tion, to hemostasis, to long-lasting depot delivery systems, to topical
`agents for systemic delivery through the skin.1–5 More relevant to this
`review are nanoparticle formulations that are delivered intravenously,
`
`and in this realm, organic nanoparticles predominantly fall into two
`categories: (a) nanoparticles for gene therapy applications22,23 or (b)
`nanoparticles for delivery of small molecule drugs for cancer treatment
`(e.g., head and neck, melanoma, breast, metastatic, etc.).24,25 Organic
`nanoparticle formulations for other applications (e.g., vaccines, fungal
`treatments, etc.) are also in development and will be highlighted here
`(Figure 1).
`The main reasons behind the interest in nanoparticle technologies
`
`are that: (a) in the case of organic nanoparticles, they possess distinct
`
`advantages over many intravenously administered pharmaceuticals
`
`and biologics, and (b) in the case of inorganic nanoparticles, many
`
`

`

`12 |
`
`stimuli responsive functions are possible based on specific colloidal
`
`assemblies. Organic nanoparticles can be designed and formulated to
`
`offer enhanced drug protection, controlled release, extended circula-
`
`tion, and improved targeting to diseased tissues as compared to their
`free drug counterparts.25,26 Likewise, inorganic nanoparticles benefit
`from these same advantages, and additionally from stimuli-responsive
`
`functions arising from their surface plasmon resonance (e.g., thermal
`
`heating or imaging) or magnetic responsiveness (e.g., magnetic reso-
`
`nance imaging [MRI] imaging or magnetic targeting) that individual
`drugs or other molecules (e.g., noncolloidal) do not offer.2,27 Given
`these advantages, it has been a long-held idea that nanoparticles have
`
`the potential to dramatically change clinical care by introducing new,
`
`or improving upon current, therapies. A large portion of the interest in
`
`nanoparticles stems from their potential as a platform delivery system,
`
`with the capability of exchanging specific design features (e.g., target-
`
`ing antibodies, the encapsulated drug, and control over how/when the
`diseased site interacts with this drug) in a “plug-and-play” format to
`treat additional or other diseases.
`
`3 | C L I N I C A L L Y A P P R O V E D
`N A N O P A R T I C L E S / MI C R O P A R T I C L E S
`
`Currently, there are a number of nanoparticle therapeutics,
`
`imaging
`
`agents, and technologies that have been approved for clinical use, either
`
`by the FDA in the United States, or the European Medicines Agency
`
`(EMA) in the European Union (Table 1). In this section, we will highlight
`
`the currently approved nanoparticles and their clinical indications.
`
`3.1 | Cancer nanoparticle medicines
`
`Many clinically approved nanoparticle formulations are used in treat-
`
`ing various cancers at a variety of stages. Interestingly, all but one of
`
`these systems (Abraxane) is liposomal systems encapsulating an anti-
`
`cancer drug. Doxil, polyethylene glycol (PEG) functionalized liposomal
`
`doxorubicin, was the first approved (FDA 1995) cancer nanomedi-
`cine.8 Soon after, other liposomal formulations such as liposomal dau-
`norubicin (DaunoXome),28 liposomal vincristine (Marqibo),29 and most
`recently liposomal irinotecan (Onivyde)30 were approved by the FDA,
`whereas non-PEGylated liposomal doxorubicin (Myocet)31 and liposo-
`mal mifamurtide (MEPACT)32 were approved by the EMA. The lone
`nonliposomal nanoparticle system currently approved for cancer treat-
`
`ments is Abraxane, which is an albumin-bound paclitaxel nanopar-
`ticle.33 The majority of these formulations are not PEGylated, with the
`exception of Doxil and Onivyde,34 which is perhaps surprising given
`the widely known advantages even small amounts of PEG have shown
`to confer to nanoparticle delivery systems.35–37 Additionally, all of
`these formulations are passively targeted, with no active or chemical-
`
`based targeting moieties; again, this is despite the proven advantages
`of active-targeting in preclinical settings.25,26,38 It is likely that the
`other advantages, notably their reduced toxicity stemming from their
`
`ability to preferentially accumulate at tumor sites and limit off-target
`side effects via the enhanced permeation and retention (EPR) effect,39
`
`ANSELMO AND MITRAGOTRI
`
`are responsible for the success and increased efficacy that these
`approved particles have over their free drug counterparts.
`
`3.2 | Iron-replacement nanoparticle therapies
`
`Another clinical area where nanoparticles have made a significant
`impact is in iron-replacement therapies for treatment of anemia (Table
`1).40–42 In these applications, the nanoparticle (iron-oxide colloids) is
`the therapeutic with the goal being to increase iron concentration in
`the body.43 These nanoparticle approaches originated from the need
`to address toxicity issues associated with the injection of
`free
`iron.40,42 Using colloidal iron coated with sugars, many of these toxic-
`ity issues were resolved.40,42 It should be noted that nanoparticles
`indicated for iron-replacement undergo vastly different approval pro-
`cedures, by both the FDA and EMA, as they are nonbiological complex
`drugs; it is a widely held belief that additional factors, stemming from
`their colloidal and nanoparticle nature, need to be considered during
`their approval (e.g., manufacturing conditions).41,44
`
`3.3 | Nanoparticle/microparticle imaging agents
`
`Alongside colloid-based iron-replacement therapies, similar iron-oxide
`nanoparticles are clinically approved as contrast agents for MRI (Table
`1).45,46 For imaging applications, the innate magnetic responsiveness
`of iron-oxide nanoparticles is used with MRI to generate contrast for
`imaging a variety of cancers and pathologies.47,48 The combination of
`an iron-oxide nanoparticle’s MRI responsiveness and small size, which
`facilitates preferential uptake in tumors, provides accurate and precise
`imaging of cancerous tissues. Interestingly, the majority of colloidal
`iron-oxide imaging agents have been discontinued in the United States
`and most of Europe.13 In addition to MRI contrast enhancers, particles
`can be used as intravenous ultrasound enhancing agents. In these
`cases, particles typically take the form of micron-sized microbub-
`bles.49,50 These microbubbles provide a means to enhance contrast by
`stabilizing and encapsulating air bubbles, which are near-perfect
`reflectors of ultrasound and would otherwise rapidly dissolve in blood
`if not encapsulated/formulated.49 Few of these products are approved
`and currently used in the clinic, for example, Definity (FDA approved)
`and SonoVue (EMA approved) are fluorocarbons or sulfur hexafluoride
`encased in lipid shells, respectively. Optison (FDA and EMA approved)
`is another ultrasound contrast agent formulated as human serum albu-
`min encased perflutren.
`
`3.4 | Nanoparticles for vaccines, anesthetics, fungal
`treatments, and macular degeneration
`
`Nanoparticles, or in these cases liposomes, are also used in a number
`of other clinical applications (Table 1). The first of these is Diprivan,51
`which was FDA approved in 1989 as a general anesthetic.52 Two vac-
`cines, Epaxal for vaccination against hepatitis A53 and Inflexal V for
`vaccination against influenza,54 are liposomal systems that have been
`approved in many European countries. Interestingly, these two vac-
`cines use their viral glycoprotein-liposomal template as the primary
`adjuvant,55 with Epaxal doing so in lieu of traditional adjuvants such as
`
`

`

`ANSELMO AND MITRAGOTRI
`
`TABLE 1 Clinically approved intravenous nanoparticle therapies and diagnostics, grouped by their broad indication
`
`Name
`
`Particle type/drug
`
`Approved application/indication
`
`Approval (year)
`
`Investigated application/indication
`
`ClinicalTrials.gov identifier
`
`Cancer Nanoparticle Medicines
`
`Doxil/Caelyx
`(Janssen)
`
`Liposomal doxorubicin
`(PEGylated)
`
`DaunoXome (Galen)
`
`Liposomal daunorubicin
`(non-PEGylated)
`
`Ovarian cancer (secondary to platinum
`based therapies)
`HIV-associated Kaposi’s sarcoma
`(secondary to chemotherapy)
`Multiple myeloma (secondary)
`HIV-associated Kaposi’s sarcoma
`(primary)
`
`FDA (1995)
`EMA (1996)
`
`Various cancers including: solid
`malignancies, ovarian, breast, leukemia,
`lymphomas, prostate, metastatic, or liver
`
`166 studies mention Doxil
`90 studies mention CAELYX
`
`FDA (1996)
`
`Various leukemias
`
`32 studies mention
`DaunoXome
`
`32 studies mention Myocet
`
`295 studies mention Abraxane
`
`Myocet (Teva UK)
`
`Abraxane (Celgene)
`
`Liposomal doxorubicin
`(non-PEGylated)
`
`Treatment of metastatic breast cancer
`(primary)
`
`Albumin-particle bound
`paclitaxel
`
`Advanced nonsmall cell lung cancer
`(surgery or radiation is not an option)
`Metastatic breast cancer (secondary)
`Metastatic pancreatic cancer (primary)
`
`EMA (2000)
`
`FDA (2005)
`EMA (2008)
`
`Various cancers including: breast,
`lymphoma, or ovarian
`
`Various cancers including: solid
`malignancies, breast, lymphomas, bladder,
`lung, pancreatic, head and neck, prostate,
`melanoma, or liver
`
`Marqibo (Spectrum)
`
`Liposomal vincristine
`(non-PEGylated)
`
`Philadelphia chromosome-negative
`acute lymphoblastic leukemia (tertiary)
`
`FDA (2012)
`
`Various cancers including: lymphoma,
`brain, leukemia, or melanoma
`
`23 studies mention Marqibo
`
`MEPACT (Millennium)
`
`Liposomal mifamurtide
`(non-PEGylated)
`
`Treatment for osteosarcoma (primary
`following surgery)
`
`EMA (2009)
`
`Osteosarcomas
`
`Onivyde MM-398
`(Merrimack)
`
`Liposomal irinotecan
`(PEGylated)
`
`Metastatic pancreatic cancer
`(secondary)
`
`FDA (2015)
`
`Various cancers including: solid
`malignancies, breast, pancreatic, sarcomas,
`or brain
`
`4 studies mention MEPACT: 3
`active/recruiting
`
`7 studies mention MM-398/
`Onivyde: 6 active/recruiting
`
`Iron-replacement nanoparticle therapies
`
`CosmoFer/INFeD/
`Ferrisat
`(Pharmacosmos)
`
`DexFerrum/DexIron
`(American Regent)
`
`Iron dextran colloid
`
`Iron deficient anemia
`
`FDA (1992)
`Some of Europe
`
`Iron deficient anemia
`
`6 studies mention INFeD:
`1 recruiting
`
`Iron dextran colloid
`
`Iron deficient anemia
`
`FDA (1996)
`
`Iron deficient anemia
`
`6 studies mention DexFerrum
`
`Ferrlecit (Sanofi)
`
`Iron gluconate colloid
`
`Iron replacement for anemia treatment
`in patients with chronic kidney disease
`
`FDA (1999)
`
`Iron deficient anemia
`
`Iron sucrose colloid
`
`Iron replacement for anemia treatment
`in patients with chronic kidney disease
`
`FDA (2000)
`
`Iron deficient anemia
`Following autologous stem cell
`transplantation
`
`13 studies mention Ferrlecit: 2
`recruiting
`
`44 studies mention Venofer
`
`Iron polyglucose
`sorbitol
`carboxymethylether
`colloid
`
`Iron deficiency in patients with chronic
`kidney disease
`
`FDA (2009)
`
`Iron deficient anemia
`Imaging: brain metastases, lymph node
`metastases, neuroinflammation in
`epilepsy, head and neck cancer,
`myocardial infarction, or multiple sclerosis
`
`57 studies mention Ferumoxytol:
`6 recruiting/active for anemia
`treatment
`22 recruiting/active for imaging
`applications
`
`Iron deficient anemia
`
`FDA (2013)
`
`Iron deficient anemia
`
`Treating iron deficiency and anemia
`when oral methods do not work or
`when iron delivery is required
`immediately
`
`Some of Europe
`
`Iron deficient anemia
`
`50 studies mention Ferinject
`8 studies mention Injectafer
`
`22 studies: 3 active/recruiting
`
`| 13
`
`Iron deficient anemia
`
`Some of Europe
`
`Iron deficient anemia
`
`1 recruiting study
`
`Injectafter/Ferinject
`(Vifor)
`
`Iron carboxymaltose
`colloid
`
`Monofer
`(Pharmacosmos)
`
`10% Iron isomaltoside
`1000 colloid
`
`Diafer
`(Pharmacosmos)
`
`5% Iron isomaltoside
`1000 colloid
`
`Venofer (American
`Regent)
`
`Feraheme (AMAG)/
`Rienso (Takeda)/
`Ferumoxytol
`
`

`

`14 |
`
`ANSELMO AND MITRAGOTRI
`
`TABLE 1 (Continued)
`
`Name
`
`Particle type/drug
`
`Approved application/indication
`
`Approval (year)
`
`Investigated application/indication
`
`ClinicalTrials.gov identifier
`
`Nanoparticle/microparticle imaging agents
`
`Definity (Lantheus
`Medical Imaging)
`
`Perflutren lipid
`microspheres
`
`Ultrasound contrast agent
`
`FDA (2001)
`
`Feridex I.V. (AMAG)/
`Endorem
`
`Iron dextran colloid
`
`Imaging of liver lesions
`
`Optison (GE
`Healthcare)
`
`Human serum albumin
`stabilized perflutren
`microspheres
`
`Ultrasound contrast agent
`
`FDA (1996)
`Discontinued
`(2008)
`
`FDA (1997)
`EMA (1998)
`
`SonoVue (Bracco
`Imaging)
`
`Phospholipid stabilized
`microbubble
`
`Ultrasound contrast agent
`
`EMA (2001)
`
`Resovist (Bayer
`Schering Pharma)/
`Cliavist
`
`Ferumoxtran-10/
`Combidex/Sinerem
`(AMAG)
`
`Nanoparticle vaccines
`
`Epaxal (Crucell)
`
`Inflexal V (Crucell)
`
`Particle anesthetics
`
`Iron carboxydextran
`colloid
`
`Imaging of liver lesions
`
`Iron dextran colloid
`
`Imaging lymph node metastases
`
`Liposome with hepatitis
`A virus
`
`Liposome with
`trivalent-influenza
`
`Hepatitis A vaccine
`
`Influenza vaccine
`
`Diprivan
`
`Liposomal propofol
`
`Induction and maintenance of
`sedation or anesthesia
`
`Some of Europe
`Discontinued
`(2009)
`
`Only available in
`Holland
`
`Some of Europe
`(Discontinued)
`
`Some of Europe
`(Discontinued)
`
`FDA (1989)
`
`Ultrasound enhancement for: liver or
`breast or intraocular or pancreatic tumors,
`pulmonary diseases, heart function,
`transcranial injuries, strokes, or liver
`cirrhosis
`
`N/A: No current studies
`
`Ultrasound enhancement for: lymph node,
`renal cell carcinoma, myocardial
`infarction, pulmonary transit times, or
`heart transplant rejections
`
`Ultrasound enhancement for: liver
`neoplasms, prostate or breast or
`pancreatic cancer, or coronary/pulmonary
`disease
`
`N/A
`No current studies
`
`Imaging lymph node metastases
`
`58 studies mention Definity
`
`4 studies mention Endorem
`2 studies mention Feridex
`No current active or recruiting
`studies
`
`11 currently active or
`recruiting studies
`
`43 studies mention SonoVue
`
`2 studies mention Resovist: No
`current active or recruiting
`studies
`
`11 studies mention
`ferumoxtran-10: 1 active
`
`Safety and immunogenicity of hepatitis A
`vaccine
`
`6 studies mention Epaxal: 1
`recruiting
`
`Safety and immunogenicity of influenza
`vaccine
`
`14 studies mention Inflexal V:
`All completed
`
`General anesthesia in specific situations:
`morbidly obese patients, open heart
`surgery, or spinal surgery
`
`110 studies mention Diprivan
`
`Nanoparticles for fungal treatments
`
`AmBisome (Gilead
`Sciences)
`
`Liposomal
`amphotericin B
`
`Cryptococcal Meningitis in HIV-
`infected patients
`Aspergillus, Candida, and/or
`Cryptococcus species infections
`(secondary)
`Visceral leishmaniasis parasite in
`immunocompromised patients
`
`FDA (1997) Most
`of Europe
`
`Preventing or treating invasive fungal
`infections
`
`50 studies mention AmBisome
`
`Nanoparticles for macular degeneration
`
`Visudyne (Bausch and
`Lomb)
`
`Liposomal verteporfin
`
`Treatment of subfoveal choroidal
`neovascularization from age-related
`macular degeneration, pathologic, or
`ocular histoplasmosis
`
`FDA (2000)
`EMA (2000)
`
`Macular degeneration
`
`52 studies mention Visudyne
`
`

`

`ANSELMO AND MITRAGOTRI
`
`aluminum hydroxide.53 However, these vaccines have since been
`phased out of the clinic. In other applications, liposomes or lipid-based
`
`nanoformulations have been clinically approved for fungal and para-
`
`sitic infections. For example, the highly toxic antifungal drug ampho-
`
`infections, has been
`tericin B, used for treating systemic fungal
`formulated in liposomes (AmBisome).56 In doing so, toxicity is dramati-
`cally reduced as the pharmacokinetics and tissue distribution is
`
`improved via liposomal encapsulation. Furthermore, the liposomal for-
`
`mulation addresses a significant issue of the free drug form of ampho-
`
`tericin B, which is its insolubility in pH 7 saline. While not true
`
`liposomes, other FDA approved lipid-complexed formulations of
`amphotericin B exist, such as Abelcet and Amphotec.57 Visudyne® is
`a light-activated liposomal
`formulation of verteporfin. Liposomal
`
`encapsulation offers enhanced uptake in proliferating cells which par-
`
`ticularly enhances targeting and subsequent uptake by targets neovas-
`
`cular
`
`areas, which,
`
`following
`
`light
`
`stimulation damages
`
`the
`
`endothelium and blocks local blood vessels to prevent and treat
`neovascularization.58
`
`4 | C U R R E N T N A N O PA R T I C L E /
`M I C R O P A R T IC L E C L I N I C A L T R I AL S
`
`Given the successes of many of these formulations in the clinic and
`
`commercial realm, significant efforts continue to explore currently
`
`approved nanomedicines as well as developing new ones. Here, we
`
`will: (a) briefly review the current clinical trial landscape for currently
`
`approved nanoparticles (Table 1), (b) review the current clinical trial
`
`landscape regarding cutting-edge nanoparticle formulations which are
`
`seeking approval (Table 2), and (c) highlight key technologies attempt-
`
`ing to integrate targeting and stimuli-responsive functions into nano-
`
`particle delivery systems.
`
`4.1 | Previously approved nanoparticles
`
`By seeking approval for additional
`
`indications, currently approved
`
`nanoparticle systems experience a more direct path to clinical approval
`
`as compared to a newer, developing, technology. This is because
`
`already approved nanoparticles have proven their safety and efficacy
`
`in humans and,
`
`if commercialized,
`
`likely meet good manufacturing
`
`practice (GMP) standards.
`
`4.1.1 | Cancer nanoparticle medicine
`
`| 15
`
`4.1.2 | Iron-replacement nanoparticle therapies
`
`Of all the FDA approved iron-replacement nanoparticle therapies, only
`
`few remain active in clinical trials. For example, CosmoFer/INFeD/Fer-
`
`risat, DexFerrum/DexIron, Ferrlecit, Monofer, and Diafer show limited
`
`activity in current clinical trials, whereas Ferinject/Injectafer, Fera-
`
`heme/Rienso/Ferumoxytol, and Venofer show dramatically more
`
`activity, mostly for iron-replacement in various clinical settings. Special
`
`attention should be placed on ferumoxytol/Feraheme/Rienso, as addi-
`
`tional approval is being sought for a number of imaging applications
`
`which is beyond its approved indication of iron-replacement (dis-
`
`cussed in detail in the next section).
`
`4.1.3 | Nanoparticle/microparticle imaging agents
`
`FDA or EMA approved iron-oxide contrast agents all show extremely
`
`low activity in current clinical trials. As stated earlier, Feridex I.V./
`
`Endorem, Resovist/Cliavist, and Combidex/Sinerem were all discontin-
`
`ued which is reflected by their lack of presence in current clinical trials.
`
`It is unlikely that these approved products will resurface in the clinic
`
`given that the manufacturer no longer produces them, either for clini-
`
`cal or research purposes. However, ferumoxytol (Feraheme or Rienso),
`
`which is approved for iron-replacement therapies is broadly investi-
`
`gated for imaging applications in the clinic. Indeed, ferumoxytol is the
`
`most widely investigated iron-oxide particle with the majority of clini-
`
`cal trials focused on imaging of various cancers or other pathologies
`
`(22 for imaging vs. 6 for anemia treatment). This is likely because there
`
`is a severe unmet need of iron-oxide imaging agents in the clinical,
`
`stemming from the discontinuation of all other iron-oxide imaging
`
`products. Approval of an iron-oxide formulation that is already used in
`
`the clinic and also mass-produced, is likely a more straight-forward
`
`path to approval as opposed to a nonapproved technology. The ultra-
`
`sound contrast enhancers SonoVue, Optison, and Definity are all being
`
`investigated in a number of clinical trials: 43, 11 active/recruiting, and
`
`58, respectively. While not a currently approved indication, except for
`
`SonoVue, few of these current clinical trials are investigating micro-
`
`bubble use for tumor imaging applications.
`
`4.1.4 | Nanoparticles for vaccines, anesthetics, fungal
`treatments, and macular degeneration
`
`Epaxal and Inflexal V, approved in some European countries as
`
`liposomal-based vaccines, are not investigated in current clinical studies,
`
`likely because they have been phased out of clinical use. In addition, the
`
`As cancer nanomedicines were approved by the FDA over 20 years
`
`platform of intravenous virosomes developed by Crucell does not
`
`ago, it is not surprising that these currently approved nanoparticles are
`
`appear to be in any current clinical trials, for any vaccine. FDA-approved
`
`investigated in the largest number of current clinical trials. For exam-
`
`Visudyne, approved for treating neovascularization is currently being
`
`ple, Doxil and Abraxane are mentioned in over 160 and 290 clinical
`
`investigated in clinical trials focused on combining it with other neovas-
`
`studies, respectively. More recently approved products such as Mar-
`
`cularization therapies. Diprivan, FDA approved in 1989, still persists in
`
`qibo, MEPACT, and Onivyde, also have a strong presence in clinical tri-
`als. These trials build on each individual nanoparticle’s current
`indications by seeking approval for: (a) additional cancer types, (b) a
`
`clinical trials, mostly for approval as an anesthetic for special cases (e.g.,
`
`morbidly obese patients, spinal or open-heart surgeries). AmBisome,
`
`approved nearly two decades ago in 1997 by the FDA, is still studied in
`
`combination therapy with other therapeutic agents, or (c) upgrading
`
`the clinic for additional bacterial/fungal infections and in tolerability and
`
`their use from a secondary therapy to a primary first-line therapy.
`
`efficacy in patients with other diseases or complications.
`
`

`

`16 |
`
`ANSELMO AND MITRAGOTRI
`
`Intravenous nanoparticle therapies and diagnostics which have not been clinically approved and are currently undergoing clinical tri-
`TABLE 2
`als (not yet recruiting, recruiting, or active), grouped by particle type as well as well as application
`
`Name (company)
`
`Particle type/drug
`
`Investigated application/indication
`
`ClinicalTrials.gov
`identifier (phase)
`
`Pegylated liposomal mitomycin-C
`
`Solid tumors
`
`NCT01705002 (Ph I)
`
`Liposomes (cancer)
`
`PROMITIL (Lipomedix
`Pharmaceuticals)
`
`ThermoDox® (Celsion)
`
`Lyso-thermosensitive liposomal
`doxorubicin
`
`Temperature-triggered doxorubicin
`release:
`Breast cancer recurrence at chest wall
`(microwave hypothermia)
`Hepatocellular carcinoma
`(radiofrequency ablation)
`Liver tumors (mild hypothermia)
`Refractory solid tumors (magnetic
`resonance high intensity focused
`ultrasound)
`
`VYEXOS CPX-351 (Celator
`Pharmaceuticals)
`
`Liposomal formulation of cytarabine:
`daunorubicin (5:1 molar ratio)
`
`Leukemias
`
`Oncoprex (Genprex)
`
`FUS1 (TUSC2) encapsulated liposome
`
`Lung cancer
`
`Halaven E7389-LF (Eisai)
`188Re-BMEDA-liposome
`
`Mitoxantrone Hydrochloride
`Liposome (CSPC ZhongQi
`Pharmaceutical Technology)
`
`Liposomal eribulin mesylate
`188Re-N,N-bis (2-mercaptoethyl)-N0,N0-
`diethylethylenediamine pegylated
`liposome
`
`Solid tumors
`
`Advanced solid tumors
`
`Mitoxantrone liposome
`
`Lymphoma and breast cancer
`
`JVRS-100
`
`Cationic liposome incorporating
`plasmid DNA complex for immune
`system stimulation
`
`Leukemia
`
`NCT00826085 (Ph I/II)
`NCT02112656 (Ph III)
`NCT02181075 (Ph I)
`NCT02536183 (Ph I)
`
`NCT01804101 (Not
`Provided)
`NCT02286726 (Ph II)
`NCT02019069 (Ph II)
`NCT01943682 (Ph I)
`NCT02269579 (Ph II)
`NCT02533115 (Ph IV)
`NCT01696084 (Ph III)
`
`NCT01455389 (Ph I/II)
`
`NCT01945710 (Ph I)
`
`NCT02271516 (Ph I)
`
`NCT02131688 (Ph I)
`NCT02596373 (Ph II)
`NCT02595242 (Ph I)
`NCT02597387 (Ph II)
`NCT02597153 (Ph II)
`
`NCT00860522 (Ph I)
`
`Lipocurc (SignPath Pharma)
`
`Liposomal curcumin
`
`Solid tumors
`
`NCT02138955 (Ph I/II)
`
`LiPlaCis (LiPlasome Pharma)
`
`MM-302 (Merrimack
`Pharmaceuticals)
`
`LIPUSU® (Nanjing Luye
`Sike Pharmaceutical Co.,
`Ltd.)
`
`Liposomal formulated cisplatin with
`specific degradation-controlled drug
`release via phospholipase A2 (PLA2)
`
`HER2-targeted liposomal doxorubicin
`(PEGylated)
`
`Paclitaxel Liposome
`
`Liposomes (gene therapy: cancer)
`
`TKM-080301 (Arbutus
`Biopharma)
`
`Lipid particle targeting polo-like kinase
`1 (PLK1) for delivery of siRNA
`
`Advanced or refractory tumors
`
`NCT01861496 (Ph I)
`
`Breast cancer
`
`Advanced solid tumors, or gastric,
`breast cancer
`
`NCT01304797 (Ph I)
`NCT02213744 (Ph II/III)
`
`NCT01994031 (Ph IV)
`NCT02142790 (Ph IV)
`NCT02163291 (Ph II)
`NCT02142010 (Not
`Provided)
`
`Hepatocellular carcinoma
`
`NCT02191878 (Ph I/II)
`
`siRNA-EphA2-DOPC
`
`siRNA liposome for EphA2 knockdown
`
`Solid tumors
`
`PNT2258 (ProNAi
`Therapeutics)
`
`BP1001 (Bio-Path Holdings)
`
`Proprietary single-stranded DNAi
`(PNT100) encapsulated in lipid
`nanoparticles
`
`Growth factor receptor bound protein-
`2 (Grb-2) antisense oligonucleotide
`encapsulated in neutral liposomes
`
`Lymphomas
`
`Leukemias
`
`DCR-MYC (Dicerna
`Pharmaceuticals)
`
`Atu027 (Silence
`Therapeutics GmbH)
`
`DsiRNA lipid nanoparticle for NYC
`oncogene silencing
`
`Solid tumors, multiple myeloma,
`lymphoma, or hepatocellular carcinoma
`
`AtuRNAi liposomal formulation for
`PKN3 knockdown in vascular
`endothelium
`
`Pancreatic cancer
`
`NCT01591356 (Ph I)
`
`NCT02378038 (Ph II)
`NCT02226965 (Ph II)
`NCT01733238 (Ph II)
`
`NCT01159028 (Ph I)
`
`NCT02110563 (Ph I)
`NCT02314052 (Ph I/II)
`
`NCT01808638 (Ph I/II)
`
`

`

`ANSELMO AND MITRAGOTRI
`
`TABLE 2 (Continued)
`
`Name (company)
`
`Particle type/drug
`
`Investigated application/indication
`
`SGT-53 (SynerGene
`Therapeutics)
`
`Cationic liposome with anti-transferrin
`receptor antibody, encapsulating
`Wildtype p53 sequence
`
`Glioblastoma, solid tumors, or
`pancreatic cancer
`
`| 17
`
`ClinicalTrials.gov
`identifier (phase)
`
`NCT02354547 (Ph I)
`NCT00470613 (Ph I)
`NCT02354547 (Ph I)
`NCT02340156 (Ph II)
`
`NCT01517464 (Ph I)
`
`NCT01829971 (Ph I)
`
`NCT02369198 (Ph I)
`
`Solid tumors
`
`Liver cancer
`
`Mesothelioma and nonsmall cell lung
`cancer
`
`SGT-94 (SynerGene
`Therapeutics)
`
`MRX34 (Mirna
`Therapeutics)
`
`TargomiRs (EnGeneIC)
`
`Liposomes (gene therapy: other)
`
`ND-L02-s0201 (Nitto
`Denko)
`
`ARB-001467 TKM-HBV
`(Arbutus Biopharma)
`
`Patisiran ALN-TTR02
`(Alnylam Pharmaceuticals)
`
`Liposomes (other)
`
`CAL02 (Combioxin SA)
`
`Nanocort (Enceladus in
`collaboration with Sun
`Pharma Global)
`
`RGI-2001 (Regimmune)
`
`RB94 plasmid DNA in a liposome with
`anti-transferrin receptor antibody
`
`Double-stranded RNA mimic of miR-
`34 encapsulated in liposomes
`
`Anti-EGFR bispecific antibody minicells
`(bacteria derived nanoparticles) with a
`miR-16 based microRNA payload
`
`siRNA lipid nanoparticle conjugated to
`Vitamin A
`
`Lipid particle containing three RNAi
`therapeutics that target three sites on
`the HBV genome
`
`Lipid nanoparticle RNAi for the
`knockdown of disease-causing TTR
`protein
`
`Sphingomyelin and cholesterol
`liposomes for toxin neutralization
`
`Liposomal Prednisolone (PEGylated)
`
`Liposomal formulaton of a-GalCer
`
`Sonazoid
`
`F-butane encapsulated in a lipid shell
`
`Polymeric and micelles (cancer)
`
`AZD2811 (AstraZeneca
`with BIND Therapeutics)
`
`BIND-014 (BIND
`Therapeutics)
`
`Aurora B kinase inhibit

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


Or .

Accessing this document will incur an additional charge of $.

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

Accept $ Charge
throbber

Still Working On It

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

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

throbber

A few More Minutes ... Still Working

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

Thank you for your continued patience.

This document could not be displayed.

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

Your account does not support viewing this document.

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

Your account does not support viewing this document.

Set your membership status to view this document.

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

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

Become a Member

One Moment Please

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

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

Your document is on its way!

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

Sealed Document

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

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


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket