7194_C008

Embed Size (px)

DESCRIPTION

cancer tech

Citation preview

  • 8.1 INTRODUCTION

    In the last deca

    nologies has ge

    prevention, diag

    to as nanomedic

    systems, incorp

    and/or improv

    systems.1 The

    large; these pro

    xo

    y

    )containing do

    (e.g., actinom

    late) (PECAq 2006 by Taylor & Frade, remarkable scientific progress in the development of novel, nanoscale tech-

    nerated rising expectations for this enabling technology to advance our efforts in the

    nosis and treatment of human disease. These nanotechnologies, commonly referred

    ines when applied to the treatment, diagnosis, monitoring, and control of biological

    orate a multitude of diverse and often unique nanosized products including new

    ed therapeutic drugs, diagnostic/surgical devices, and targeted drug delivery

    list of nanoparticle medicines approved and/or currently being developed is quite

    ducts range from early forms of drug-encapsulating liposomes (e.g., liposomes

    rubicin for treatment of breast and ovarian cancer) and simple polymeric structures

    cin D adsorbed on poly(methylcryanoacrylate) (PMCA) and poly(ethylcryanoacry-

    nanosized particles)2 to more intricate multifunctional nanoplatforms and8 Nanotechnology: RegulatoryPerspective for DrugDevelopment in CancerTherapeutics

    N. Sadrieh and T. J. Miller

    CONTENTS

    8.1 Introduction ......................................................................................................................... 139

    8.2 FDA Experience with Nanotechnology Product Applications........................................... 140

    8.3 FDA Definition of Nanotechnology ................................................................................... 143

    8.4 FDA Initiatives in Nanotechnology .................................................................................... 143

    8.5 Research at the FDA on Nanotechnology .......................................................................... 145

    8.6 Regulatory Considerations for Nanotechnology Products ................................................. 145

    8.6.1 Jurisdiction of Nanotechnology Products ............................................................... 145

    8.6.2 Existing FDA Regulations That Apply to Nanomaterial-Containing Products ..... 146

    8.7 Scientific Considerations for the Development of Nanotechnology Products .................. 147

    8.7.1 Characterization of Nanoparticles........................................................................... 147

    8.7.2 Safety Considerations.............................................................................................. 149

    8.8 Conclusions ......................................................................................................................... 149

    8.9 Trademark Listing ............................................................................................................... 151

    References .................................................................................................................................... 151139

    ncis Group, LLC

  • developed to improve early detection of cancer and/or to provide alternative clinical therapeutic

    approaches to fighting cancer, some of these products have been approved by the Food and Drug

    Administration for clinical use.75

    A a-

    teria is

    avail r-

    gane ct

    the r es

    inclu i-

    bition of oxidative stress and lipid peroxidation in isolated microsomes, dermal fibroblasts, andcultured cortical neurons;7779 cytotoxicity of metal oxide nanoparticles to BRL 3A liver cells;80

    and macrophage apoptosis after exposure to carbon nanomaterials and ultrafine particles.8183

    Although a vast amount of useful information has been obtained from these various in vitro

    models, the relevance and application of these findings to human drug safety remain uncertain

    until validated with in vivo studies with identical nanomaterials.

    Targeted in vivo efficacy data is readily available for many of the novel, potential nanother-

    apeutics; however, necessary in vivo data indicating mechanism of action, general

    pharmacology/toxicity profiles, and optimal product characterization efforts rarely appear in the

    published literature. Much of our current understanding of the biocompatibility of nanomaterials

    comes from focused inhalation and dermal studies using nanosized products and ultrafine contami-

    nants, as a model of environmental and occupational risk.8487 However, there are questions

    regarding the level of applicability of these studies to human drug safety assessment due to the

    variability in size and purity of the chemical or structural composition reported or not reported in

    these studies, the route of primary exposure, and the complex differences in the structure and

    function of the affected organs.88 In addition, other in vivo studies, which identify intrinsic anti-

    oxidant and oxidative stress-inducing properties of unsubstituted fullerenes in a rodent and fish

    species, respectively, have important but limited value in drug discovery in the absence of systemic,

    whole animal toxicology data67,89.

    While the enthusiasm and expectations for success in utilizing nanomaterials and nanometer-

    sized structures in biomedical applications grow, so do the concerns regarding the potential impact

    of such products on the environment and human health.88 The absence of available safety infor-

    mation for a vast majority of these novel nanosized materials and nanoparticlized drugs in

    biological systems only further supports the need for additional research into any potential toxic

    mechanisms in humans. The questions that appear most often in deciding the safety of any nanos-

    cale material include:

    Are nanomaterials and nanotechnology new to the United States Food and DrugAdministration (FDA)?

    How do existing regulations ensure the development of safe and effective nanotech-nology-based drugs and how is the FDA preparing to deal with nanotechnology?

    What are the scientific considerations that raise unique concerns for nanotechnology-based therapeutics?

    8.2 FDA EXPERIENCE WITH NANOTECHNOLOGY PRODUCT APPLICATIONSHisto

    char

    q 2006lthough there have been many articles published on the in vitro toxicity of certain nanom

    ls, very little in vivo safety data evaluating the general toxicity of these novel products

    able in the published literature. Many in vitro assays (cell-based, organelle-based, and cell/o

    lle-free systems) have been used to evaluate the cytotoxicity of nanomaterials to help predi

    elative safety and general biocompatibility of the nanoproducts in vivo. Several such studi

    de viability assays in vascular endothelial cells exposed to C60;76 fullerene induction/inhdiagnostic devices, including polymeric dendrimers, nanocrystal quantum dots, carbon-based full-

    erenes, and nanoshells, to name a few (see Table 8.1). The vast majority of these products are being

    Nanotechnology for Cancer Therapy140rically, the FDA has encountered and approved many products with particulate materials

    acteristic of a nanomedicine or nanoproduct (e.g., small size, mechanism of delivery, increased

    by Taylor & Francis Group, LLC

  • TABLE 8.1Nanomedicines: Drugs/Devices FDA Approved and in Development

    Name Structure Company Size (nm) Phase Indication Referencea,b

    Magnevistwc PIO Shering AG !1 APP MRI tumor imaging 35

    Feridexwc SPIO Adv.Magnetics 120180 APP MRI tumor imaging 59

    Rapamunewc Nanocrystal drug Wyeth 1001,000 APP Immunosuppressant agent 1012

    Emendwc Nanocrystal drug Merck 1001,000 APP Nausea 13,14

    Doxilwc Liposome-drug encapsulation Ortho Biotech z100 APP Metastatic ovarian cancer 1517

    Abraxanewc Nanoparticulate albumin American Pharmaceutical

    Partners

    z130 APP Non-small-cell lung cancer, breast cancer,

    others

    1820

    SilvaGardw Silver nanoparticles AcryMed z10 APP Antimicrobial surface treatment (medical

    devices)

    2123

    AmBisomewc Liposome-drug encapsulation Gilead Science 4580 APP Fungal infections 2426

    Leunessew Solid lipid nanoparticles Aano-therapeutics N/A On market Cosmetics N/A

    NB-001d Nanoemulsion NanoBio Corp z150 Phase II Topical microbicide for herpes labialis 2730

    VivaGelw Dendrimer Starpharma Holdings Ltd. N/A Phase I Vaginal microbicide (STI and HIV

    prevention)

    3134

    INGN-401c Drugliposome complex Introgen Therapeutics N/A Phase I Metastatic lung cancer 3538

    Combidexwc USPIO Advanced Magnetics 2050 NDA filed MRI tumor imaging 3942

    IT-101c Drugpolymer complex Insert Therapeutics N/A IND filed Metastatic solid tumors 43,44

    Dendrimersc Polymeric spheres, multifunctional Dendritech Nanotech.,

    Starpharma

    O1 to !500 IDV Diagnostic, contrast agents, microbicide,

    drug delivery, B neutron capture

    4554

    Nanoshellsc Metal shell, SiO2 core Nanospectra 100200 IDV Photo-thermal ablation therapy /imaging

    tumors

    5558

    Fullerenesc Carbon sphere, C60, multifunctional C Sixty 1 IDV MRI contrast agent, antiviral, antioxidant 5967

    Quantum dotsc CdSea nanocrystal Evident Technologies 210 IDV Optical imaging, tumor imaging,

    photosensitizer

    6874

    APP, approved; IDV, in development; PIO, paramagnetic iron oxide; SPIO, superparamagnetic iron oxide; USPIO, ultrasmall superparamagnetic iron oxide; STI, sexually transmitted

    infection; HIV, human immunodeficiency virus; N/A, not available.

    a Most typical semiconductor metal used, however can also be composed of nearly any other semiconductor metal (e.g., CdS, CdTe, ZnS, PbS, etc.).b References do not necessarily refer to the product described, and may reflect general properties about the product.c Drugs with proposed or proven diagnostic or therapeutic benefit to cancer patients.d Unable to locate peer-reviewed publications. References listed are press releases found during internet search.

    q 2006 by Taylor & Francis Group, LLC

    Regu

    latory

    Persp

    ectivein

    Nan

    otech

    nolo

    gyPro

    ducts

    141

  • surface area, specialized function related to size and increased surface area, etc.). Although the field

    of nanomedicine may be new, the submission of applications for products containing novel dosage

    forms or drug delivery systems, including nanomaterials, nanoparticlized drugs and medical

    devices, is not particularly new to this agency. Products are reviewed on a product-by-product

    basis, and as such, various concepts of risk management (i.e., risk identification, risk analysis, risk

    control, etc.) are often implemented to support the drug review process.

    The FDA is aware of several FDA regulated products that employ nanotechnology. However,

    to date, few manufacturers of regulated products have claimed the use of nanotechnology in the

    manufacture of their products or made any nanotechnology claims for the finished product.

    The FDA is aware that a few cosmetic products and sunscreens claim to contain nanoparticles

    to increase the product stability, modify release of ingredients, or change the opacity of the

    product by using nanoparticles of titanium dioxide and zinc oxide. Similarly, several drugs

    such as Gd-DTPA (Magnevistw, tumor contrast agent),35 ferumoxides (Feridexw, tumor contrast

    agent),59 liposomal doxorubicin (Doxilw, chemotherapeutic),1517 and albumin-bound paclitaxel

    (Abraxanew, chemotherapeutic),1820 to name a few, have all been approved for human clinical

    use in the diagnosis and treatment of a variety of patient tumors and metastatic cancers. For public

    information regarding FDA approval letters, label information, and review packages for each

    of these drugs and others listed in Table 8.1, please visit the following website: http://www.

    accessdata.fda.gov/scripts/cder/drugsatfda/.

    Of the many drugs listed in Table 8.1, tumor contrast agents comprise some of the earliest

    nanosized products to receive FDA approval for human clinical use. Critical to therapeutic

    outcome, diagnostic imaging of nanosized contrast agents using non-invasive magnetic resonance

    imaging (MRI) and positron emission tomography (PET), have shown advanced utility for early

    detection of small tumors, metastatic lymph nodes, and altered tumor microenvironments.90

    The leading MR contrast agent, Magnevistw, a paramagnetic, gadolinium-based contrast

    medium, and Feridexw, a superparamagnetic iron oxide contrast agent, received primary FDA

    approval for clinical use in 1988 and 1996, respectively. Each contrast agent has a different

    mean particle size (!1 nm and 120180 nm, respectively), is approved for different clinical indi-cations, and demonstrates product-specific adverse sensitivity and pharmacology/toxicology

    profiles in patients.39

    Another early nanomedicine, liposomal encapsulated doxorubicin (Doxilw), is regulated by the

    FDA and has been available in the clinic for treatment of various cancers since 1995. Drug

    incorporation inside the hydrophilic core or within the hydrophobic phospholipid bilayer coat of

    liposomes, has been shown to improve drug solubility, enhance drug transfer into cells and tissues,

    facilitate organ avoidance, and modify drug release profiles, minimizing toxicity.91 The liposomal

    formulation of the popular anthracyline, doxorubicin, which is commonly used to treat metastatic

    breast and ovarian cancer, is reported to have diminished cardiotoxicity and enhanced therapeutic

    efficacy compared to the free form of the drug.1517 This increased efficacy is most likely due to the

    passive targeting of solid tumors through the enhanced permeability and retention effect inherent to

    tumor vasculature and aberrant tumor morphology.17 The approximate diameter of the doxoru-

    bicinliposomal product is reported to be 100 nm, near the size limits described in the FDA

    definition of a nanomedicine.

    Nanoparticlized, albumin-bound paclitaxel, Abraxanew, is one of the first FDA-approved

    (January, 2005) cancer drugs that was submitted to the FDA with specific claims regarding

    nano-related characteristics that improved this particular formulation over previous sub-

    missions.1820 Initially identified as (ABI-007), this chemotherapeutic agent that is approved for

    the treatment of metastatic breast cancer eliminates the excipient Cremophorw by overcoming

    intrinsic insolubility with nanoparticlization of the active paclitaxel ingredient. Although widely

    used in many drug formulations, Cremophor has been associated with an adverse hypersensitivity20

    Nanotechnology for Cancer Therapy142reaction that limited the amount of paclitaxel that could be safely administered. Unlike the

    previous three agents, the approximate diameter of Abraxane is 130 nm, slightly larger than the

    q 2006 by Taylor & Francis Group, LLC

  • technology is defined as the following: In case of these devices, nanoscale objects were defined as

    molecules or devices within the size range of one to hundreds of nanometers that are the active

    component or object, even within the frame-work of a larger micro-size device or at a macro-

    need to use medicines and foods to improve their health. Towards this end, the FDA needs to

    proactively pursue those innovations that would result in a significant enhancement of publichealth. Therefore, it falls directly within the mandate of its mission for the FDA to understand

    and help to speed innovations that promise to improve many products that will be regulated by the

    FDA, such as those resulting from nanotechnology.

    In response to a slowdown in innovating medical therapies submitted to the FDA for approval, the

    FDA released the Critical Path Initiative (http://www.fda.gov/oc/initiatives/criticalpath/whitepaper.interface. A discussion regarding the definition of nanotechnology is currently an active topic

    among various organizations. However, while a definition is always useful, the FDA has tradition-

    ally dealt with products on a case-by-case basis. This approach is likely to continue for

    nanotechnology products, regardless of the definition. However, we are actively involved in the

    various organizations and committees dealing with issues regarding terminology, such as the

    American Society for Testing and Materials (ASTM) International.

    The definition of nanotechnology at the FDA has been consistent with the definition of nano-

    technology as reported by the NNI. As such, the FDA currently defines nanotechnology with the

    following definition:

    1. The existence of materials or products at the atomic, molecular, or macromolecular

    levels, where at least one dimension that affects the functional behavior of the drug/de-

    vice product is in the length scale range of approximately 1100 nm.

    2. The creation and use of structures, devices, and systems that have novel properties and

    functions because of their small size.

    3. The ability to control or manipulate the product on the atomic scale.

    8.4 FDA INITIATIVES IN NANOTECHNOLOGY

    The FDA is responsible for protecting the health of the public by assuring the safety, efficacy and

    security of human and veterinary drugs, biological products, medical devices, our nations food

    supply, cosmetics, and products that emit radiation. The FDA is also responsible for advancing the

    publics health by helping to speed innovations that make medicines and foods more effective, safer

    and more affordable; and by helping the public get the accurate, science-based information theyrequirements described in the National Nanotechnology Initiative (NNI) definition of nanotech-

    nology (!100 nm), and thus originally was determined not to fit the size requirement of ananomedicine. However, the fact that the exact particle size exceeds 100 nm may not necessarily

    preclude Abraxane and other similarly-sized or larger products from being

    considered nanotechnology.

    8.3 FDA DEFINITION OF NANOTECHNOLOGY

    As described on the NNI website (http://www.nano.gov/index.html), Nanotechnology is the

    understanding and control of matter at dimensions of roughly 1100 nm, where unique phenomena

    enable novel applications. Encompassing nanoscale science, engineering and technology, nano-

    technology involves imaging, measuring, modeling, and manipulating matter at this length scale.

    Similarly, in a recent publication by the European Science Foundation on Nanomedicine,92 nano-

    Regulatory Perspective in Nanotechnology Products 143html) in March 2004. The report describes the urgent need to modernize the medical product

    development processthe Critical Pathto make product development more predictable and less

    q 2006 by Taylor & Francis Group, LLC

  • costly. The Critical Path Initiative at the FDA is a serious attempt to bring attention and focus to the

    need for targeted scientific efforts to modernize the techniques and methods used to evaluate the

    safety, efficacy, and quality of medical products as they move from product selection and design to

    mass manufacture. It is intended to integrate new science into the regulatory process. As such, a list of

    opportunities for the Critical Path Initiative was published by the FDA, and nanotechnology was

    identified as one of the elements under the Critical Path Initiative. Figure 8.1 illustrates the three

    dimensions of the Critical Path (safety, medical utility and industrialization).

    However, the Food and Drug Administration is only one of several government agencies that

    currently regulates and evaluates a wide range of products that utilize nanotechnology and/or

    contain nanomaterials, including foods, cosmetics, drugs, devices, and veterinary products. As a

    member of both the National Science and Technology Council (NSTC) Committee on Technology,

    and the Nanoscale Science, Engineering and Technology (NSET) Working Group on Nanomater-

    ials Environmental and Health Implications (NEHI), the FDA coordinates with other government

    agencies, including the National Institute for Environmental Health Sciences (NIEHS), the

    National Toxicology Program (NTP), and the National Institute of Occupational Safety and

    Health (NIOSH) to develop novel strategies for the characterization of and safety evaluation

    standards for nanomaterials. The FDA also works closely with the National Cancer Institute

    (NCI) and the National Institute of Standards and Technology (NIST) in the Interagency Oncology

    Task Force, Nanotechnology subcommittee, to pool knowledge and resources to facilitate the

    development and approval of new cancer drugs and to address the use of nanotechnology in

    cancer therapies and diagnostics. Within the FDA, the Office of Science and Health Coordination

    manages the regular interaction and discussion of nanotechnology between the various agency

    centers and offices, and the nanotechnology working groups within each center, and address the

    Nanotechnology for Cancer Therapy144concerns about the regulation of nanosized drugs and materials submitted to the individual centers.

    Basicresearch

    Safety

    Medicalutility

    Industrial-ization

    Dim

    ensi

    ons

    Prototypedesign ordiscovery

    Material selectionstructureactivity

    relationship

    In vitroand animal

    testing

    Humanand animal

    testing

    In vitro andcomputer

    modelevaluation

    In vitroand animal

    models

    Humanefficacy

    evaluation

    Physicaldesign

    Characterizationsmall-scaleproduction

    Manunfacturingscale-uprefined

    specifications

    Massproduction

    Safetyfollow

    up

    Preclinicaldevelopment Clinical development

    FDA filling/approval &

    launchpreparation

    FIGURE 8.1 A highly generalized description of activities that must be successfully completed at different pointsand in different dimensions along the Critical Path. Many of these activities are highly complexwhole industries

    are devoted to supporting them. Not all the described activities are performed for every product, and many activitieshave been omitted for the sake of simplicity. (From http://www.fda.gov/oc/initiatives/criticalpath/whitepaper.

    html.)

    q 2006 by Taylor & Francis Group, LLC

  • which happen to be conducted by the Center for Drug Evaluation and Research (CDER), include:1. Particle size determination in marketed sunscreens with TiO2 and ZnO nanoparticles.

    Sunscreens are considered drugs in the U.S., and although some undergo a premarket

    FDA review, others are marketed under over-the-counter (OTC) monographs that outline

    the active ingredients that the FDA has found to be safe and effective, as well as labeling

    that is truthful and not misleading. TiO2 and ZnO used in sunscreens are therefore

    manufactured according to OTC monographs, and these monographs do not specify

    particle size. As a result, a manufacturer can formulate a sunscreen using particles of

    TiO2 and ZnO that may or may not be in the nano size range. Therefore, to assess the

    particle size of TiO2 and ZnO in currently marketed sunscreens, CDER has undertaken a

    research project, in collaboration with NIST.

    2. Another project focuses on manufacturing various nanoformulations in our laboratories

    and characterizing the physical and chemical properties of the nanoparticles in these

    formulations, including evaluating the effects of excipients on the measured parameters,

    the effects of preparation methodology on the measured parameters, and the impact (if

    any) that process and formulation variables may have on nanotechnology product

    characteristics and stability.

    3. Another project is studying the in vivo effects of selected nanoparticles, using validated

    animal models. The toxicity of the same nanoparticles will also be evaluated, using

    various in vitro systems.

    8.6 REGULATORY CONSIDERATIONS FOR NANOTECHNOLOGY PRODUCTS

    8.6.1 JURISDICTION OF NANOTECHNOLOGY PRODUCTS

    There has been much interest in the question of whether nanotechnology products will be

    considered drugs, devices, biologics or a combination of the three for regulatory purposes and

    assignment of work. This has been extensively discussed internally within the FDA and the current

    thinking is that many of these products will be considered combination products.

    Combination products (i.e., drugdevice, drugbiologic, and devicebiologic products) are

    increasingly incorporating cutting edge, novel technologies that hold great promise for advancing

    patient care. For example, innovative drug delivery devices have the potential to make treatments

    safer, more effective, or more convenient to patients. For example, drug-eluting cardiovascular

    stents have the potential to reduce the need for surgery by preventing the restenosis that sometimes8.5 RESEARCH AT THE FDA ON NANOTECHNOLOGY

    Although the FDA is a regulatory agency, it is also engaged in a number of research activities.

    However, the FDA recognizes that science supported by research provides the foundation for sound

    regulation. As such, the FDA is interested in research projects that can address specific regulatory

    needs. As an example, FDA has a grants program in support of orphan products research and

    development. The FDA does not have a grants program to support other research in non-FDA

    laboratories. Nevertheless, in several of its Centers the FDA conducts research to understand the

    characteristics of nanomaterials and nanotechnology processes. Research interests include any

    areas related to the use of nanoproducts that the FDA needs to consider in the regulation of

    these products.

    Although many FDA Centers are engaged in some nanotechnology research activities, only

    some of the specific projects that are currently ongoing are listed below. The research projects,

    Regulatory Perspective in Nanotechnology Products 145occurs following stent implantation. Drugs and biologics can be used in combination to potentially

    enhance the safety and/or effectiveness of either product used alone. Biologics are being

    q 2006 by Taylor & Francis Group, LLC

  • which they are assigned.A combination product is assigned to an agency center or alternative organizational component

    that will have primary jurisdiction for its premarket review and regulation. Under section 503(g)(1)

    of the act, assignment to a center with primary jurisdiction, or a lead center, is based on a

    determination of the primary mode of action (PMOA) of the combination product. For

    example, if the PMOA of a devicebiological combination product is attributable to the biological

    product, the agency component responsible for premarket review of that biological product would

    have primary jurisdiction for the combination product.

    A final rule defining the PMOA of a combination product was published in the August 25, 2005

    edition of the Federal Register, and is available at http://www.fda.gov/oc/combination/. The final

    rule defines PMOA as the single mode of action of a combination product that provides the most

    important therapeutic action of the combination product.

    8.6.2 EXISTING FDA REGULATIONS THAT APPLY TO NANOMATERIAL-CONTAINING PRODUCTS

    The FDA has specific guidance/requirements in place for most products. These guidance docu-

    ments can be accessed on the FDAs website (http://www.fda.gov/oc/industry/default.htm). These

    requirements apply to all products, whether they contain nanomaterials or not. Existing require-

    ments are therefore considered to be adequate at this time for most nanotechnology

    pharmaceutical products.

    In the following section, there will be a brief description of the current preclinical requirements

    for the submission of most drugs, to highlight why we believe that our current regulations are

    adequate for the evaluation of the types of nanotechnology products that have been reported as

    being close to submission to the agency as an investigational new drug (IND).

    Within CDER, the preclinical requirements for approval to market pharmaceutical products

    include both short-term and long-term toxicity testing in rodent and non-rodent species. Speci-

    fically, the types of studies conducted by pharmaceutical manufacturers prior to New Drug

    Application (NDA) submission include pharmacology (mechanism of action), safety pharmacology

    (functional studies in various organ systems, most notably the cardiovascular system), absorption,

    distribution, metabolism, and excretion (ADME), genotoxicity (potential to cause mutations in both

    in vivo and in vitro assays), developmental toxicity (to assess effects on reproduction, fertility andincorporated into novel orthopedic implants to help facilitate the regeneration of bone required to

    permanently stabilize the implants.

    Because combination products involve components that would normally be regulated under

    different types of regulatory authorities, and frequently by different FDA centers, they also raise

    challenging regulatory, policy, and review management issues. A number of criticisms have been

    raised regarding the FDAs regulation of combination products. These include concerns about the

    consistency, predictability, and transparency of the assignment process; issues related to the

    management of the review process when two (or more) FDA centers have review responsibilities

    for a combination product; lack of clarity about the postmarket regulatory controls applicable to

    combination products; and lack of clarity regarding certain Agency policies, such as when appli-

    cations to more than one agency center are needed.

    To address these concerns, FDAs Office of Combination Products (OCP) was established on

    December 24, 2002, as required by the Medical Device User Fee and Modernization Act of 2002

    (MDUFMA). The law gives the office broad responsibilities covering the regulatory life cycle of

    drugdevice, drugbiologic, and devicebiologic combination products. However, the primary

    regulatory responsibilities for, and oversight of, specific combination products will remain in

    one of three product centersthe Center for Drug Evaluation and Research, the Center for

    Biologics Evaluation and Research, or the Center for Devices and Radiological Healthto

    Nanotechnology for Cancer Therapy146lactation), irritation studies (to assess local irritation effects), immunotoxicology (to assess effects

    on the components of the immune system), carcinogenicity (to assess the capacity of drugs to

    q 2006 by Taylor & Francis Group, LLC

  • nology applications that it is only possible to assess the safety of nanomaterials when the materialunder study is adequately characterized. If two or more laboratories are working on what they think

    is the same material, then in order for the data from one laboratory to be compared with data from

    another laboratory, there needs to be some confirmation that in fact the nanomaterials being studied

    are actually identical. It has been mentioned by many that for nanomaterials, small differences in

    product characteristics (such as size, surface charge, hydrophobicity, or a number of other attri-

    butes) may impact product behavior and thus result in vastly different safety profiles. Published data

    show that slight modifications of a nanoparticle have resulted in different toxicity profiles. For

    example, several in vitro studies with Starburstw polyamidoamine (PAMAM) dendrimers and

    Caco-2 cells have shown decreased cytotoxicity of cationic dendrimers upon addition of lauroyl

    or polyethylene glycol (PEG) to the surface of the macromolecules.9394 Similar findings of dimin-

    ished toxicity were observed in J774 macrophages upon conjugation of polysaccharides to polymer

    nanoparticles and in fibroblasts and CHO cells with the addition of PEGsilica to the surface of

    CdSe and CdSe/ZnS nanocrystals.9596

    Although much less toxicity data is available in vivo, it is evident that surface chemistry is of

    similar importance to the toxico- and pharmacokinetics, toxico- and pharmacodynamics, and9799induce tumors in animal models) and other possible studies (specific studies for a product being

    developed). The current battery of tests, as listed above, is therefore considered to be adequate

    because of the following reasons: the studies use high dose multiples of the drug (usually three

    doses, one that results in low, one that results in medium and one that results in high toxicity), at

    least two animal species are used (usually one rodent and one non-rodent), extensive histopathology

    is conducted on most organs (where most organs are removed, fixed, stained and viewed under the

    microscope to assess if there are structural changes that may have resulted from damage to the

    organs), functional tests are conducted to assess if there are effects on specific organ systems (such

    as the heart, brain, respiratory system, reproductive system, immune system, etc.) and drug treat-

    ments in animals can be for extended periods of time (up to two years for carcinogenicity studies).

    Additional studies might be requested based on drug-specific considerations and on a case-

    by-case basis. Therefore, as for other drugs, nanotechnology products will be handled on a case-

    by-case basis, depending on the characteristics of the particular product being developed. As

    research provides additional understanding with regards to the toxicity of nanomaterials, the

    FDA may require additional toxicological tests to ensure the safety of the products it regulates.

    8.7 SCIENTIFIC CONSIDERATIONS FOR THE DEVELOPMENT

    OF NANOTECHNOLOGY PRODUCTS

    Ensuring the safety of nanomaterials in drug applications is one of the most important concerns for

    regulators and drug developers. However, as important as safety is, it is also important to

    adequately characterize the nanomaterials used in drug products. Proper characterization and

    manufacturing procedures will ensure that a consistent product is being used, both during precli-

    nical and clinical development, and after approval. Some of the questions that have been raised

    internally regarding the assurance of safety of nanomaterial-containing products, as well as ques-

    tions regarding characterization, are listed below. It is not within the scope of this chapter to address

    questions regarding manufacturing and scale-up; however, it should be noted that this is an issue

    that needs serious consideration, because unique characteristics of nanotechnology products are

    likely to require unique manufacturing procedures.

    8.7.1 CHARACTERIZATION OF NANOPARTICLES

    It has become clear to most of those involved in research or development of potential nanotech-

    Regulatory Perspective in Nanotechnology Products 147overall stability of the nanomaterials in vivo. When injected into rodents, several generations

    of dendrimers with different surface configurations displayed contrasting organ distribution

    q 2006 by Taylor & Francis Group, LLC

  • patterns and plasma clearance profiles in vivo, typically depositing within the liver, kidney, and

    pancreas.98100 PEG-conjugation to the dendrimer particle surface significantly increased the blood

    half-life and diminished the liver accumulation of dendrimer particles.99 Similarly, quantum dots

    coated with a complex polymer with carbon alkyl side chains demonstrated greater in vivo stability

    and less genotoxicity than those coated with a simpler polymer or lipid coating.101 Based on the

    findings of these studies and the collection of pulmonary inhalation data with environmental

    ultrafines and other in vitro cytotoxicity results obtained with different nanomaterials (e.g.,

    quantum dots, fullerenes, and nanoshells, etc.), it appears that a thorough characterization of the

    nanomaterial is essential to the interpretation and understanding of toxicity data collected from

    in vitro and in vivo studies.

    To summarize some of the ongoing discussions within CDERs nanotechnology working

    group, a list of questions has been put together regarding the characterization of nanomaterial-

    containing products. At this time, it appears that there are no adequate answers to these questions.

    However, it may be that in the future, to satisfy regulatory requirements for nanomaterial-

    containing products, the questions listed below will need to be addressed by product sponsors.

    1. What are the forms in which particles are presented to the organism, the cells and the

    organelles? Are these particles soluble or insoluble, are the nanomaterials organic or

    inorganic molecules, are the nanomaterials described as nanoemulsions, nanocrystal

    colloid dispersions, liposomes, nanoparticles that are combination products (drug

    device, drugbiologic, drugdevicebiologic), or something else? Does the product

    fall within the currently established definition of nanotechnology?

    2. What are the tools that can be used to characterize the properties of the nanoparticles?

    Can standard tools that are used of other types of products (for example, spectroscopic

    tools) be used to characterize nanomaterial-containing products, or do these products

    require specialized tools that are not widely available for product characterization

    (atomic force microscopy, tunneling electron microscopy, or other specialized modal-

    ities)?

    3. Are there validated assays to detect and quantify nanoparticles in the drug product and in

    tissues?

    4. How can long- and short-term stability of nanomaterials be assessed? Specifically, how

    can the stability of nanomaterials be assessed in various environments (buffer, blood,

    plasma, and tissues)?

    5. What are the critical physical and chemical properties of nanomaterials in a drug product

    and how do residual solvents, processing variables, impurities and excipients impact

    these properties?

    6. How do physical characteristics impact product quality and performance? Within what

    range can these physical properties be modified without significantly affecting product

    quality and performance?

    7. What are the critical steps in the scale-up and manufacturing process for nanotechnology

    products? How could manufacturing procedures for nanomaterials differ from those for

    other types of drugs?

    8. How are issues concerning the characterization and manufacturing procedures for nano-

    technology products assessed, when considering the development of personalized

    therapies? In this context, personalized therapies refer to those multifunctional products

    that will be custom made for specific patients, depending on issues such as the

    expression of a certain receptor on a tumor or the response of the tumor to a particular

    chemotherapeutic agent. For these types of therapies, what should be the level of

    Nanotechnology for Cancer Therapy148characterization of the nanomaterials? Does the preclinical testing need to be repeated

    for each modification of the multifunctional molecule, or can there be limited testing to

    q 2006 by Taylor & Francis Group, LLC

  • 8.7.2 SAFETY CONSIDERATIONS

    Other than characterization, the other major consideration has to do with safety. However, it is only

    within the context of adequately characterized products that safety studies provide value. There-

    8.8 CONCLUSIONS

    In this chapter, we have tried to highlight some of the steps being taken by the FDA to meet the

    ch r-

    ou al

    wo as

    ini er

    un

    typ

    q 2allenges of nanotechnology products. The FDA has engaged in an open discussion with nume

    s groups and organizations, has participated in and sponsored workshops, has established intern

    rking groups, has outlined a path for the review of combination nanotechnology products and h

    tiated research projects in nanotechnology. All these efforts have been geared towards a bettfore, although safety is instinctively the primary concern, adequate characterization of

    nanomaterials has to precede any studies on safety.

    As particle size decreases, there may be size-specific effects. These effects may impact safety if

    the materials are more reactive. It has been mentioned in numerous discussions that, as particle size

    decreases, reactivity is expected to rise. The increased reactivity combined with the decreased size

    has led us to pose many questions. Some of these questions are listed below:

    1. Will nanoparticles gain access to tissues and cells that normally would be bypassed by

    larger particles?

    2. If nanoparticles enter cells, what effects do they have on cellular and tissue functions?

    Are the effects transient or permanent? Might there be different effects in different cell

    types, depending on specific tissue targets or receptors?

    3. Once nanoparticles enter tissues, how long do they remain there and where do they

    concentrate?

    4. What are the mechanisms by which nanoparticles are cleared from tissues and blood and

    how can their clearance be measured?

    5. What are some route-specific issues for nanomaterials, and how are these issues different

    than for other types of materials that are not in the nanoscale?

    6. With regards to ADME, specific questions include:

    a. What are the differences in the ADME profile for nanoparticles versus larger particles

    of the same drug?

    b. Are current methods used for measuring drug levels in blood and tissues adequate for

    assessing levels of nanoparticles (appropriateness of method, limits of detection)?

    c. How accurate are mass balance studies, especially if the levels of drug administered

    are very low; i.e., can 100% of the amount of the administered drug be accounted for?

    d. How is clearance of targeted nanoparticles accurately assessed?

    e. If nanoparticles concentrate in a particular tissue, how can clearance be accurately

    determined?

    f. Can nanoparticles be successfully labeled for ADME studies?cover certain aspects of the product behavior, such as the ADME, the toxicology (a

    bridging study for the acute and/or repeat dose toxicology studies)? What aspects of

    the chemistry, manufacturing and controls (CMC) studies need to be repeated and what

    should be the extent of physical characterization?

    Regulatory Perspective in Nanotechnology Products 149derstanding of the science and towards assessing the adequacy of existing regulations for the

    es of products that are expected to be regulated by the FDA.

    006 by Taylor & Francis Group, LLC

  • At this point, and based on internal discussions, the FDA feels that its current regulations are

    adequate to address the types of nanotechnology products that are being developed as drugs or drug

    delivery systems. The reasons for this have been outlined in this chapter. Although there have been

    requests by investigators and developers of nanotechnology products for the FDA to issue nano-

    technology-specific guidances, no new guidance documents applicable specifically to

    nanotechnology products will be issued at this time. This is because it is felt that all existing

    Guidance documents apply to these products. All products being developed, whether nanotech-

    nology-based or not, are unique and therefore likely to have unique issues requiring consideration.

    As such, and as for all other products reviewed by the FDA, nanotechnology products will be

    reviewed on a case-by-case basis.

    As shown in Figure 8.2, the drug development process is multiphasic and along the path, there

    are numerous opportunities for meetings between the sponsor and the FDA. The first meeting is the

    pre-IND meeting, and it is highly recommended that sponsors of nanotechnology products take the

    opportunity to meet with the FDA at this early stage. An early meeting with the FDA can avoid

    unnecessary or unfocussed studies that could result in a waste of sponsor resources. During early

    meetings, sponsors can present, with minimal data, their preclinical plans to the FDA and obtain

    guidance on how to focus the studies submitted in the IND package. This type of interaction

    between the FDA and the sponsor will be valuable to both parties and will pave the way for the

    most efficient drug development plan.

    Basicresearch

    Prototypedesign ordiscovery

    Pre-IND meetingIndustry - FDA End of Safety update

    Preclinicaldevelopment

    Clinical development FDA filing/approval &

    launchpreparationPhase 1 Phase 2 Phase 3

    Nanotechnology for Cancer Therapy150interactionsduringdevelopment Initial

    INDsubmissions

    Ongoingsubmission

    IND review phase Applicationreviewphase

    Pre-BLA or NDAmeeting

    End of phase2 meeting

    phase2a meeting

    Marketapplicationsubmission

    FIGURE 8.2 This figure depicts the extensive industryFDA interactions that occur during product develop-ment, using the drug development process as a specific example.* Developers often meet with the agency

    before submitting an investigational new drug (IND) application to discuss early development plans. An IND

    must be filed and cleared by the FDA before human testing can commence in the United States. During the

    clinical phase, there are ongoing submissions of new protocols and results of testing. Developers often request

    additional meetings to get FDA agreement on the methods proposed for evaluation of safety or efficacy, and

    also on manufacturing issues. *Note: Clinical drug development is conventionally divided into three phases.This is not the case for medical device development. (From www.fda.gov/oc/initiatives/criticalpath/white-

    paper.html.)

    q 2006 by Taylor & Francis Group, LLC

  • superparamagnetic iron oxide MR contrast agents and commonly used transfection agents, Academy

    of Radiology, 9 (Suppl. 2), S484S487, 2002.7. Shapiro, E. M., Skrtic, S., Sharer, K., Hill, J. M., Dunbar, C. E., and Koretsky, A. P., MRI detection of

    single particles for cellular imaging, Proceedings of the National Academy of Sciences of the United

    States of America, 101 (30), 1090110906, 2004.

    8. Daldrup-Link, H. E., Rudelius, M., Oostendorp, R. A., Settles, M., Piontek, G., Metz, S.,

    Rosenbrock, H., Keller, U., Heinzmann, U., Rummeny, E. J., Schlegel, J., and Link, T. M., Targeting

    of hematopoietic progenitor cells with MR contrast agents, Radiology, 228 (3), 760767, 2003.

    9. Kostura, L., Kraitchman, D. L., Macka, A. M., Pittenger, M. F., and Bulte, J. W., Feridex labeling of

    mesenchymal stem cells inhibits chondrogenesis but not adipogenesis or osteogenesis, NMR in

    Biomedicine, 17 (7), 513517, 2004.

    10. LaVan, D. A., Lynn, D. M., and Langer, R., Moving smaller in drug discovery and delivery, National

    Review of Drug Discovery, 1, 7784, 2002.

    11. Miller, J. L., Sirolimus approved with renal transplant indication, American Journal of Health-

    System Pharmacy, 56 (21), 21772178, 1999.

    12. Aspeslet, L. J. and Yatscoff, R. W., Requirements for therapeutic drug monitoring of sirolimus, an

    immunosuppressive agents used in renal transplant, Clinical Therapeutics, 22 (Suppl. B), B86B92,

    2000.8.9 TRADEMARK LISTING

    1. Magnevistw (Schering Aktiengesellschaft, Berlin, Germany)

    2. Feridexw (Advanced Magnetics, Inc., Cambridge, Massachusetts, U.S.A.)

    3. Rapamunew (Wyeth, Madison, New Jersey, U.S.A.)

    4. Emendw (Merck & CO., Inc., Whitehouse Station, New Jersey, U.S.A.)

    5. Doxilw (Ortho Biotech Products, L.P., Bridgewater, New Jersey, U.S.A.)

    6. Abraxanew (American Pharmaceutical Partners, Inc., Schaumburg, Illinois, U.S.A.)

    7. SilvaGardw (AcryMed, Beaverton, Oregon, U.S.A.)

    8. AmBisomew (Gilead Sciences, Inc., Foster City, California, U.S.A.)

    9. Leunessew (Nanotherapeutics, Inc., Alachua, Florida, U.S.A.)

    10. VivaGelw (Starpharma Holdings, Ltd., Melbourne, Australia)

    11. Combidexw (Advanced Magnetics, Inc., Cambridge, Massachusetts, U.S.A.)

    12. Cremophorw (BASF Aktiengesellschaft, CO., Ludwigshafen, Germany)

    13. Starburstw (Dendritic Nanotechnologies, Inc., Mount Pleasant, Michigan, U.S.A.)

    REFERENCES

    1. Moghimi, S. M., Hunter, A. C., and Murray, J. C., Nanomedicine: Current status and future

    prospects, FASEB Journal, 19 (311), 330, 2005.

    2. Brigger, I., Dubernet, C., and Couvreur, P., Nanoparticles in cancer therapy and diagnosis, Advanced

    Drug Delivery Review, 54 (5), 631651, 2002.

    3. Kieffer, S. A., Gadopentetate dimeglumine: Observations on the clinical research process,

    Radiology, 174 (1), 78, 1990.

    4. Zhang, H. L., Ersoy, H., and Prince, M. R., Effects of gadopentetate dimeglumine and gadodiamide

    on serum calcium, magnesium, and creatinine measurements, Journal of Magnetic Resonance

    Imaging, 23, 383387, 2006.

    5. de Lussanet, Q. G., Backes, W. H., Griffioen, A. W., van Engleshoven, J. M., and Beets-Tan, R. G.,

    Gadopentenate dimeglumine versus ultra-small superparamagnetic iron oxide for dynamic contrast-

    enhanced MR imaging of tumor angiogenesis in human colon carcinoma in mice, Radiology, 229,

    429438, 2003.

    6. Frank, J. A., Zywicke, H., Jordan, E. K., Mitchell, J., Lewis, B. K., Miller, B., Bryant, L. H. Jr., and

    Bulte, J. W., Magnetic intracellular labeling of mammalian cells by combining (FDA approved)

    Regulatory Perspective in Nanotechnology Products 15113. Patel, L. and Lindley, C., Aprepitant-a novel NK1-receptor antagonist, Expert Opinion on Pharma-

    cotherapy, 4 (12), 22792296, 2003.

    q 2006 by Taylor & Francis Group, LLC

  • 14. Wu, Y., Loper, A., Landis, E., Hettrick, L., Novak, L., Lynn, K., Chen, C., Thompson, K., Higgins,

    R., Batra, U., Shelukar, S., Kwei, G., and Storey, D., The role of biopharmaceutics in the develop-

    ment of a clinical nanoparticle formulation of MK-0869: A beagle dog model predicts improved

    bioavailability and diminished food effect on absorption in human, International Journal of Phar-

    maceutics, 285 (12), 135146, 2004.

    15. Markman, M., Kennedy, A., Webster, K., Peterson, G., Kulp, B., and Belinson, J., Phase 2 trial of

    liposomal doxorubicin (40 mg/m(2)) in platinum/paclitaxel refractory ovarian fallopian tube cancers

    and primary carcinoma of the peritoneum, Gynecologic Oncology, 78, 369372, 2000.

    16. Frykman, G., Williams, G., and Pazdur, R., Conflicting phase II efficacy date for Doxil, Journal of

    Clinical Oncology, 19 (2), 596597, 2001.

    17. Laginha, K. M., Verwoert, S., Charrois, G. J. R., and Allen, T. M., Determination of doxorubicin

    levels in whole tumor and tumor nuclei in murine breast cancer tumors, Clinical Cancer Research, 11

    (19), 69446949, 2005.

    18. Harries, M., Ellis, P., and Harper, P., Nanoparticle albumin-bound paclitaxel for metastatic breast

    cancer, Journal of Clinical Oncology, 23 (31), 77687771, 2005.

    19. Nyman, D. W., Campbell, K. J., Hersh, E., Long, K., Richardson, K., Trieu, V., Desai, N., Hawkins,

    M. J., and Von Hoff, D. D., Phase I and pharmacokinetics trial of ABI-007, a novel nanoparticle

    formulation of paclitaxel in patients with advanced nonhematologic malignancies, Journal of

    Clinical Oncology, 23 (31), 77857793, 2005.

    20. Sparreboom, A., Scripture, C. D., Trieu, V., Williams, P. J., De, T., Yang, A., Beals, B., Figg, W. D.,

    Hawkins, M., and Desai, N., Comparative preclinical and clinical pharmacokinetics of a cremaphor-

    free nanoparticle albumin-bound paclitaxel (ABI-007) and Paclitaxel formulated in cremaphor

    (Taxol), Clinical Cancer Research, 11 (11), 41364143, 2005.

    21. Chaw, K. C., Manimaran, M., and Tay, F. E., Role of silver ions in destabilization of intermolecular

    adhesion forces measured by atomic force microscopy in staphylococcus epidermis biofilms, Anti-

    microbial Agents and Chemotherapy, 49 (12), 48534859, 2005.

    22. Warriner, R. and Burrell, R., Infection and the chronic wound: A focus on silver, Advanced Skin

    Wound Care, 18 (Suppl. 1), 212, 2005.

    23. Baker, C., Pradhan, A., Pakstis, L., Pochan, D. J., and Shah, S. I., Synthesis and antibaceterial

    properties of silver nanoparticles, Journal of Nanoscience and Nanotechnology, 5 (2), 244249,

    2005.

    24. Takemoto, K., Yamamoto, Y., Ueda, Y., Sumita, Y., Yoshida, K., and Niki, Y., Comparative study

    on the efficacy of Ambisome and Fungizone in a mouse model of pulmonary aspergillosis, Journal of

    Antimicrobial Chemotherapy, 57 (4), 724731, 2006.

    25. Cetin, H., Yalaz, M., Akisu, M., Hilmioglu, S., Metin, D., and Kultursay, N., The efficacy of two

    different lipid-based amphotericin B in neonatal Candida septicemia, Pediatrics International, 47

    (6), 676680, 2005.

    26. Barratt, G. and Legrand, P., Comparison of the efficacy and pharmacology of formulations of

    amphotericin B used in treatment of leishmaniasis, Current Opinion in Infectious Diseases, 18

    (6), 527530, 2005.

    27. The Healthcare Sales and Marketing Network. NanoBiow corporation announces FDA approval for

    phase II clinical trials for herpes labialis treatment. http://salesandmarketingnetwork.com/news_-

    release.php?IDZ14361 (accessed February 24, 2004).28. NewsRX. FDA approves phase II clinical trials for herpes labialis treatment. http://www.newsrx.

    com/article.php?articleIDZ87034 (accessed February 24, 2004).29. BioSpace. NanoBio announces FDA approval for phase II clinical trials for herpes labialis treatment.

    http://www.biospace.com/news_story.aspx?StoryIDZ15257520&fullZ1 (accessed February 24,2004).

    30. Nanotsunami. NanoBiow completes successful phase 2 herpes trial, prepares for phase 3 trial. http://

    www.voyle.net/Nano%20Medicine%202005/Medicine%202005-0087.htm (accessed July 22,

    2005).

    31. Abner, S. R., Guenthner, P. C., Guarner, J., Hancock, K. A., Cummins, J. E. Jr., Fink, A., Gilmore,

    G. T., Staley, C., Ward, A., Ali, O., Binderow, S., Cohen, S., Grohskopf, L. A., Paxton, L., Hart,

    Nanotechnology for Cancer Therapy152C. E., and Dezzutti, C. S., A human colorectal explant culture to evaluate topical microbicides for the

    prevention of HIV infection, Journal of Infectious Diseases, 192 (9), 15451556, 2005.

    q 2006 by Taylor & Francis Group, LLC

  • 32. Gong, E., Matthews, B., McCarthy, T., Chu, J., Holan, G., Raff, J., and Sacks, S., Evaluation of

    dendrimer SPL7013, a lead microbicide candidate against herpes simplex virus, Antiviral Research,

    68 (3), 139146, 2005.

    33. McCarthy, T. D., Karellas, P., Henderson, S. A., Giannis, M., OKeefe, D. G., Heery, G., Paull,

    J. R. A., Matthews, B. R., and Holan, G., Dendrimers as drugs, discovery and preclinical and clinical

    development of dendrimer-based microbicides for HIV and STI prevention, Molecular Pharma-

    cology, 2 (4), 312318, 2005.

    34. Jiang, Y. H., Emau, P., Cairns, J. S., Flanary, L., Morton, W. R., McCarthy, T. D., and Tsai, C. C.,

    SPL7013 gel as topical microbicide for prevention of vaginal transmission of SHIV89.6P in maca-

    ques, AIDS Research and Human Retroviruses, 21 (3), 207213, 2005.

    35. Ito, I., Ji, L., Tanaka, F., Saito, Y., Gopalan, B., Branch, C. D., Xu, K., Atkinson, E. N., Bekele, B. N.,

    Stephens, L. C., Minna, J. D., Roth, J. A., and Ramesh, R., Liposomal vector mediated delivery of the

    3p FUS1 gene demonstrates potent antitumor activity against human lung cancer in vivo, Cancer

    Gene Therapy, 11 (11), 733739, 2004.

    36. Smalltimes. Study: Nano therapy kills tumors, extends life. http://www.smalltimes.com/document_-

    display.cfm?section_idZ53&document_idZ8429 (accessed November 11, 2004).37. Nanotechnology News. Introgens nanoparticle tumor treatment increases survival rate by 70%.

    http://www.azonano.com/news.asp?newsIDZ406 (accessed November 12, 2004).38. Nanotsunami. Introgens nanoparticle cancer therapy INGN 401 demonstrates promise in the treat-

    ment of lung cancer. http://www.voyle.net/Nano%20Medicine%202005/Medicine%202005-0052.

    htm (accessed May 17, 2005).

    39. Bourrinet, P., Bengele, H. H., Bonnemain, B., Dencausse, A., Idee, J. M., Jacobs, P. M., and Lewis,

    J. M., Preclinical safety and pharmacokinetic profile of ferumoxtran-10, an ultrasmall superpara-

    magnetic iron oxide magnetic resonance contrast agent, Investigative Radiology, 41 (3), 313324,

    2006.

    40. Saksena, M. A., Saokar, A., and Harisinghani, M. G., Lymphotropic nanoparticle enhanced MR

    imaging (LNMRI) technique for lymph node imaging, European Journal of Radiology, 58 (3),

    367374, 2006.

    41. Harisinghani, M. G., Saksena, M. A., Hahn, P. F., King, B., Kim, J., Torabi, M. T., and Weissleder,

    R., Ferumoxtran-10-enhanced MR lymphangiography: Does contrast-enhanced imaging alone

    suffice for accurate lymph node characterization?, American Journal of Roentgenology, 186 (1),

    144148, 2006.

    42. Metz, S., Lohr, S., Settles, M., Beer, A., Woertler, K., Rummeny, E. J., and Daldrup-Link, H. E.,

    Ferumoxtran-10-enhanced MR imaging of the bone marrow before and after conditioning therapy in

    patients with non-Hodgkin lymphomas, European Radiology, 16 (3), 598607, 2006.

    43. Schluep, T., Hwang, J., Cheng, J., Heidel, J. D., Barlett, D. W., Hollister, B., and Davis, M. E.,

    Preclinical efficacy of the camptothecin-polymer conjugate IT-101 in multiple cancer models,

    Clinical Cancer Research, 12 (5), 16061614, 2006.

    44. Schluep, T., Cheng, J., Khin, K. T., and Davis, M. E., Pharmacokinetics and biodistribution of the

    camptothecinpolymer conjugate IT-101 in rats and tumor-bearing mice, Cancer Chemotherapy and

    Pharmacology, 57, 654662, 2006.

    45. Newkome, G. R., Yao, Z., Baker, G. R., and Gupta, V. K., Cascade molecules: A new approach to

    micelles. A (27)-Arborol, Journal of Organic Chemistry, 50, 20032004, 1985.

    46. Tomalia, D. A., Baker, H., Dewald, J. R., Hall, M., Kallos, G., Maris, S., Roeck, J., Ryder, J., and

    Smith, P. A., New class of polymers: Starburstdendritic macromolecules, Polymer Journal (Tokyo),

    17, 117123, 1985.

    47. Wiener, E. C., Brechbiel, M. W., Brothers, H., Magin, R. L., Gansow, O. A., Tomalia, D. A., and

    Lauterbur, P. C., Dendrimer-based metal chelates: A new class of magnetic resonance imaging

    contrast agents, Magnetic Resonance in Medicine, 31 (1), 18, 1994.

    48. Grinstaff, M. W., Biodendrimers: New polymeric biomaterials for tissue engineering, Chemistry, 8,

    28392846, 2002.

    49. Bourne, N., Stanberry, L. R., Kern, E. R., Holan, G., Matthews, B., and Berstein, D. I., Dendrimers, a

    Regulatory Perspective in Nanotechnology Products 153new class of candidate topic microbicides with activity against herpes simplex virus infestation,

    Antimicrobial Agents in Chemotherapy, 44, 24712474, 2000.

    q 2006 by Taylor & Francis Group, LLC

  • Nanotechnology for Cancer Therapy15450. Zanini, D. and Roy, R., Practical synthesis of PAMAM athiosialodendrimers for probing multivalent

    carbohydratelectin binding properties, Journal of Organic Chemistry, 63, 34863491, 1998.

    51. Zhuo, R. X., Du, B., and Lu, Z. R., In vitro release of 5-fluoroacil with cyclic core dendritic polymer,

    Journal of Controlled Release, 57, 249257, 1999.

    52. Liu, M., Kono, K., and Frechet, J. M. J., Water-soluble dendritic unimolecular micelles: Their

    potential as drug delivery agents, Journal of Controlled Release, 65, 121131, 2000.

    53. Kobayashi, H. and Brechbiel, M. W., Dendrimer-based nanosized MRI contrast agents, Current

    Pharmaeutical Technology, 5, 539549, 2004.

    54. Konda, S. D., Wang, S., Brechbiel, M., and Wiener, E. C., Biodistribution of a 153 Gd-folate

    dendrimer generationZ4, in mice with folate-receptor positive and negative ovarian tumor xeno-grafts, Investigative Radiology, 37, 199204, 2002.

    55. Hirsch, L. R., Stafford, R. J., Bankson, J. A., Sershen, S. R., Rivers, B., Price, R. E., Hazle, J. D.,

    Halas, N. J., and West, J. L., Nanoshell-mediated nar-infared thermal therapy of tumors under

    magnetic resonance guidance, Proceedings of the National Academy of Sciences of the United

    States of America, 100, 1354913554, 2003.

    56. Loo, C., Lin, A., Hirsch, L., Lee, M. H., Barton, J., Halas, N., West, J., and Drezek, R., Nanoshell-

    enabled photonics-based imaging and therapy of cancer, Technology Cancer Research and Treat-

    ment, 3, 3340, 2004.

    57. Loo, C., Lowery, A., Halas, N., West, J., and Drezek, R., Immunotargeted nanoshells for integrated

    cancer imaging and therapy, Nano Letters, 5, 709711, 2005.

    58. ONeal, D. P., Hirsch, L. R., Halas, N., Payne, J. D., and West, J. L., Photo-thermal tumor ablation in

    mice using near-infrared-absorbing nanoparticles, Cancer Letters, 209 (2), 171176, 2004.

    59. de Jong, W. H., Roszek, B., and Geertsman, R. E., Nanotechnology in medical applications: Possible

    risks for human health. RIVM Report 265001002, Bilthoven, 2005.

    60. Haddon, R. C., Hebard, A. F., Rosseinsky, M. J., Murphy, D. W., Duclos, S. J., Lyons, K. B., Miller,

    B., Rosamilia, J. M., Fleming, R. M., Kortan, A. R., Glarum, S. H., Makjiha, A. V., Muller, A. J.,

    Eick, R. H., Zahurak, S. M., Tycko, R., Dabbagh, G., and Thiel, F. A., Conducting films of C60 and

    C70 by alkali-metal doping, Nature, 350, 320322, 1991.

    61. Bosi, S., Da Ros, T., Spalluto, G., and Prato, M., Fullerene derivatives: An attractive tool for

    biological applications, European Journal of Medical Chemistry, 38, 913923, 2003.

    62. Friedman, S. H., Decamp, D. L., Sijbesma, R. P., Srdanov, G., Wudl, F., and Kenyon, G. L.,

    Inhibition of the HIV-1 protease by fullerene derivatives: Model building studies and experimental

    verification, Journal of the American Chemical Society, 115, 65066509, 1993.

    63. Schiniazi, R. F., Sijbesma, R., Srdanov, G., Hill, C. L., and Wudl, F., Synthesis and virucidal activity

    of a water soluble, configurationally stable, derivatized C60 fullerene, Antimicrobial Agents and

    Chemotherapy, 37, 17071710, 1993.

    64. Chueh, S. C., Lai, K. M., Lee, M. S., Chiang, L. Y., Ho, T., and Chen, S. C., Decrease of free radical

    level in organ perfusate by a novel water soluble carbon-sixty hexa(sulfobutyl)fullerenes, Transplant

    Proceedings, 31, 19761977, 1999.

    65. Lin, A. M., Chyi, B. Y., Wang, S. D., Yu, H., Kanakamma, P. P., Luh, T. Y., Chou, C. K., and Ho,

    L. T., Carboxyfullerene prevents iron-induced oxidative stress in rat brain, Journal of Neuro-

    chemistry, 72, 16341640, 1999.

    66. Cagle, D. W., Kennel, S. J., Mirzadeh, S., Alford, J. M., and Wilson, L. J., In vivo studies of

    fullerene-based materials using endohedral metallofullerene radiotracers, Proceedings of the

    National Academy of Sciences of the United States of America, 96, 51825187, 1999.

    67. Gharbi, N., Pressac, M., Hadchouel, M., Szwarc, H., Wilson, S. R., and Moussa, F., (60)Fullerene is a

    powerful antioxidant in vivo with no acute or subacute toxicity, Nano Letters, 5 (12), 25782585,

    2005.

    68. Bruchez, M. Jr., Moronne, M., Gin, P., Weiss, S., and Alivisatos, A. P., Semiconductor nanocrystals

    as fluorescent biological labels, Science, 281, 20132016, 1998.

    69. Sukhanova, A., Venteo, L., Devy, J., Artemyev, M., Oleinikov, V., Pluot, M., and Nabiev, I., Highly

    stable fluorescent nanocrystals as a novel class of labels for immunohistochemical analysis ofparaffin embedded tissue sections, Laboratory Investigation, 82, 12591261, 2002.

    q 2006 by Taylor & Francis Group, LLC

  • 70. Sukhanova, A., Devy, J., Venteo, L., Kaplan, H., Artemyev, M., Oleinikov, V., Klinov, D., Pluot, M.,

    Cohen, J. K., and Nabiev, I., Biocompatible fluorescent nanocrystals for immunolabeling of

    membrane proteins and cells, Analytical Biochemistry, 324, 6067, 2004.

    71. Lidke, D. S., Nagy, P., Heintzmann, R., Arndt-Jovin, D. J., Post, J. N., Freco, H. E., Jares-Erijman,

    E. A., and Jovin, T. M., Quantum dot ligands provide new insights into erbB/HER receptor mediated

    signal transduction, Nature Biotechnology, 22, 198203, 2004.

    72. Akerman, M. E., Chan, W. C., Laakkonen, P., Bhatia, S. N., and Ruoslahti, E., Nanocrystal targeting

    in vivo, Proceedings of the National Academy of Sciences of the United States of America, 99,

    1261712621, 2002.

    73. Kim, S., Lim, Y. T., Soltesz, E. G., De Grand, A. M., Lee, J., Nakayama, A., Parker, J. A.,

    Mihalijevic, T., Laurence, R. G., Dor, D. M., Cohn, L. H., Bawendi, M. G., and Frangioni, J. V.,

    Near-infrared fluorescent type II quantum dots for sentinel lymph node mapping, Nature Bio-

    technology, 22, 9397, 2003.

    74. Ballou, B., Lagerholm, B. C., Ernst, L. A., Bruchez, M. P., and Waggoner, A. S., Noninvasive

    imaging of quantum dots in mice, Bioconjugate Chemistry, 15, 7986, 2004.

    75. Duncan, R., Nanomedicine gets clinical, Materials Today, 8 (8 Suppl. 1), 1617, 2005.

    76. Yamawaki, H. and Iwai, N., Cytotoxicity of water-soluble fullerene in vascular endothelial cells,

    American Journal of Physiology. Cell Physiology, 290 (6), C1495C1502, 2006.

    77. Kamat, J. P., Devasagayam, T. P. A., Priyadarsini, K. I., and Mohan, H., Reactive oxygen species

    mediated membrane damage induced by fullerene derivatives and its possible biological impli-

    cations, Toxicology, 155, 5561, 2000.

    78. Sayes, C. M., Gobin, A. M., Ausman, K. D., Mendez, J., West, J. L., and Colvin, V. L., Nano-C60

    cytotoxicity is due to lipid peroxidation, Biomaterials, 26, 75877595, 2005.

    79. Dugan, L. L., Gabrielsen, J. K., Yu, S. P., Lin, T. S., and Choi, D. W., Buckminsterfullerenol free

    radical scavengers reduce excitotoxic and apoptotic death of cultured cortical neurons, Neurobiology

    of Disease, 3, 129135, 1996.

    80. Hussain, S. M., Hess, K. L., Gearhart, J. M., Geiss, K. T., and Shlager, J. J., In vitro toxicity of

    nanoparticles in BRL 3A rat liver cells, Toxicology In Vitro, 19, 975983, 2005.

    81. Jia, F., Wang, H., Yan, L., Wang, X., Pei, R., Yan, T., Zhao, Y., and Guo, X., Cytotoxicity of carbon

    nanomaterials: Single-wall nanotube, multi-wall nanotube, and fullerene, Environmental Science &

    Technology, 39, 13781383, 2005.

    82. Renwick, L. C., Donaldson, K., and Clouter, A., Impairment of alveolar macrophage phagocytosis

    by ultrafine particles, Toxicology and Applied Pharmacology, 172, 119127, 2001.

    83. Moller, W., Hofer, T., Ziesenis, A., Karg, E., and Heyder, J., Ultrafine particles cause cytoskeletal

    dysfunctions in macrophages, Toxicology and Applied Pharmacology, 182, 197207, 2002.

    84. Oberdorster, G., Toxicology of ultrafine particles: In vivo studies, Philosophical Transactions of the

    Royal Society of London, Series A, 358, 27192740, 2000.

    85. Brown, D. M., Wilson, M. R., Macnee, W., Stone, V., and Donaldson, K., Size-dependent proin-

    flammatory effects of ultrafine polystyrene particles: A role for surface area and oxidative stress in

    the enhanced activity of ultrafines, Toxicology and Applied Pharmacology, 175, 191199, 2001.

    86. Johnston, C. H., Finkelstein, J. N., Mercer, P., Corson, N., Gelein, R., and Oberdorster, G., Pulmon-

    ary effects induced by ultrafine PTFE particles, Toxicology and Applied Pharmacology, 168,

    208215, 2000.

    87. Oberdorster, G., Oberdorster, E., and Oberdorster, J., Nanotoxicology: An emerging discipline

    evolving from studies of ultrafine particles, Environmental Health Perspectives, 113 (7), 823839,

    2005.

    88. Holsapple, M. P. and Lehman-McKeeman, L. D., Forum series: Research strategies for safety

    evaluation of nanomaterials, Toxicological Sciences, 87 (2), 315, 2005.

    89. Oberdorster, E., Manufactured nanomaterials (fullerenes, C60) induce oxidative stress in the brain of

    juvenile largemouth bass, Environmental Health Perspectives, 112 (10), 10581062, 2004.

    90. Sullivan, D. C. and Ferrari, M., Nanotechnology and tumor imaging: Seizing an opportunity, Mol-

    ecular Imaging, 3 (4), 364369, 2004.

    Regulatory Perspective in Nanotechnology Products 15591. Portney, N. G. and Ozkan, M., Nano-oncology: Drug delivery, imaging, sensing, Analytical and

    Bioanalytical Chemistry, 384, 620630, 2006.

    q 2006 by Taylor & Francis Group, LLC

  • 92. Nanomedicine, an ESF-European Medical Research Councils, (EMRC) forward look report. http://

    www.esf.org/, 2005.

    93. El-Sayed, M., Ginski, M., Rhodes, C., and Ghandehari, H., Transepithelial transport or poly(ami-

    doamine) dendrimers across Caco-2 cell monolayers, Journal of Controlled Release, 81, 355365,

    2002.

    94. Jevprasesphant, R., Penny, J., Jalal, R., Attwood, D., McKeown, N. B., and DEmanuele, A. D., The

    influence of surface modification on the cytotoxicity of PAMAM dendrimers, International Journal

    of Pharmaceutics, 252, 263266, 2003.

    95. Lemarchand, C., Gref, R., Pasirani, C., Garcion, E., Petri, B., Muller, R., Costantini, D., and

    Couvreur, P., Influence of polysaccharide coating on the interactions of nanoparticles with biological

    systems, Biomaterials, 27, 108118, 2006.

    96. Kirchner, C., Liedl, T., Kudera, S., Pellegrino, T., Javier, A. M., Gaub, H. E., Stolzle, S., Fertig, N.,

    and Parak, W. J., Cytotoxicity of colloidal CdSe and CdSe/ZnS nanoparticles, Nano Letters, 5 (2),

    331338, 2005.

    97. Neerman, M. F., Zhang, W., Parrish, A. R., and Simanek, E. E., In vitro and in vivo evaluation of a

    melamine dendrimer as a vehicle for drug delivery, International Journal of Pharmaceutics, 281,

    129132, 2004.

    98. Nigavekar, S., Sung, L. Y., Llanes, M., El-Jawahri, A., Lawrence, R. S., Becker, C. W., Balogh, L.,

    and Khan, M. K., 3H Dendrimer nanoparticle organ/tumor distribution, Pharmaceutical Research,

    21 (3), 476483, 2004.

    99. Malik, N., Wiwattanapatapee, R., Klopsch, R., Lorenz, K., Frey, H., Weener, J. W., Meijer, E. W.,

    Paulus, W., and Duncan, R., Relationship between structure and biocompatibility in vitro, and

    preliminary studies on the biodistribution of 125I-labeled polyamidoamine dendrimers in vivo,

    Journal of Controlled Release, 65, 133148, 2000.

    100. Roberts, J. C., Bhalgat, M. K., and Zera, R. T., Preliminary biological evaluation of polyamidoamine

    Nanotechnology for Cancer Therapy156(PAMAM) starburst dendrimers, Journal of Biomedical Materials Research, 30 (1), 5365, 1996.

    101. Hardman, R., A toxicological review of quantum dots: Toxicity depends on physiochemical and

    environmental factors, Environmental Health Perspectives, 114 (2), 165172, 2006.q 2006 by Taylor & Francis Group, LLC

    Table of ContentsChapter 8: Nanotechnology: Regulatory Perspective for Drug Development in Cancer Therapeutics8.1 INTRODUCTION8.2 FDA EXPERIENCE WITH NANOTECHNOLOGY PRODUCT APPLICATIONS8.3 FDA DEFINITION OF NANOTECHNOLOGY8.4 FDA INITIATIVES IN NANOTECHNOLOGY8.5 RESEARCH AT THE FDA ON NANOTECHNOLOGY8.6 REGULATORY CONSIDERATIONS FOR NANOTECHNOLOGY PRODUCTS8.6.1 JURISDICTION OF NANOTECHNOLOGY PRODUCTS8.6.2 EXISTING FDA REGULATIONS THAT APPLY TO NANOMATERIAL-CONTAINING PRODUCTS

    8.7 SCIENTIFIC CONSIDERATIONS FOR THE DEVELOPMENT OF NANOTECHNOLOGY PRODUCTS8.7.1 CHARACTERIZATION OF NANOPARTICLES8.7.2 SAFETY CONSIDERATIONS

    8.8 CONCLUSIONS8.9 TRADEMARK LISTINGREFERENCES