Title of Invention

A COMPOSITION COMPRISING A DENDRIMER COMPLEX

Abstract A composition comprising a dendrimer complex, wherein said first dendrimer is covalently linked to said second dendrimer, said dendrimer complex comprising first and second dendrimers, said first dendrimer comprising a first agent and said second dendrimer comprising a second agent, wherein said first agent is different than said second agent.
Full Text FIELD OF THE INVENTION
The present invention relates to novel therapeutic and diagnostic systems. More
particularly, the present invention is directed to dendrimer based multifunctional
compositions and systems for use in disease diagnosis and therapy (e.g., cancer diagnosis
and therapy). The compositions and systems generally comprise two or more separate
components for targeting, imaging, sensing, and/or triggering release of a therapeutic or
diagnostic material and monitoring the response to therapy of a cell or tissue (e.g., a
tumor).
BACKGROUND OF THE INVENTION
New initiatives in chemotherapeutics and radiopharmaceutics have improved the
survival of patients with many forms of neoplasm. Several cancers now have five year
survival rates greater than 80 percent. However, despite these successes, many problems
still exist concerning cancer therapy. For example, many common neoplasms, such as
colon cancer, respond poorly to available therapies.
For tumor types that are responsive to current methods, only a fraction of cancers
respond well to the therapies. In addition, despite the improvements in therapy for many
cancers, most currently used therapeutic agents have severe side effects. These side
effects often limit the usefulness of chemotherapeutic agents and result in a significant
portion of cancer patients without any therapeutic options. Other types of therapeutic
initiatives, such as gene therapy or immunotherapy, may prove to be more specific and
have fewer side effects than chemotherapy. However, while showing some progress in a
few clinical trials, the practical use of these approaches remains somewhat limited at this
time.
Despite the limited success of existing therapies, the understanding of the
underlying biology of neoplastic cells has advanced. The cellular events involved in
neoplastic transformation and altered cell growth are now identified and the multiple
steps in carcinogenesis of several human tumors have been documented (See e.g., Isaacs,
Cancer 70:1810 [1992]). Oncogenes that cause unregulated cell growth have been
identified and characterized as to genetic origin and function. Specific pathways that
regulate the cell replication cycle have been characterized in detail and the proteins
involved in this regulation have been cloned and characterized. Also, molecules that
mediate apoptosis and negatively regulate cell growth have been clarified in detail (Kerr
et al., Cancer 73:2013 [1994]). It has now been demonstrated that manipulation of these
cell regulatory pathways has been able to stop growth and induce apoptosis in neoplastic
cells (See e.g., Cohen and Tohoku, Exp. Med., 168:351 [1992] and Fujiwara et al., J.
Natl. Cancer Inst., 86:458 [1994]). The metabolic pathways that control cell growth and
replication in neoplastic cells are important therapeutic targets.
Despite these impressive accomplishments, many obstacles still exist before these
therapies can be used to treat cancer cells in vivo. For example, these therapies require
the identification of specific pathophysiologic changes in an individual's particular tumor
cells. This requires mechanical invasion (biopsy) of a tumor and diagnosis typically by in
vitro cell culture and testing. The tumor phenotype then has to be analyzed before a
therapy can be selected and implemented. Such steps are time consuming, complex, and
expensive.
There is a need for treatment methods that are selective for tumor cells compared
to normal cells. Current therapies are only relatively specific for tumor cells. Although
tumor targeting addresses this selectivity issue, it is not adequate, as most tumors do not
have unique antigens. Further, the therapy ideally should have several., different
mechanisms of action that work in parallel to prevent the selection of resistant
neoplasms, and should be releasable by the physician after verification of the location
and type of tumor. Finally, the therapy ideally should allow the physician to identify
residual or minimal disease before and immediately after treatment, and to monitor the
response to therapy. This is crucial since a few remaining cells may result in re-growth,
or worse, lead to a tumor that is resistant to therapy. Identifying residual disease at the
end of therapy (i.e., rather than after tumor regrowm) would facilitate eradication of the
few remaining tumor cells.
Thus, an ideal therapy should have the ability to target a tumor, image the extent
of the tumor and identify the presence of the therapeutic agent in the tumor cells. It
ideally allows the physician to determine why cells transformed to a neoplasm, to select
therapeutic molecules based on the pathophysiologic abnormalities in the tumor cells, to
activate the therapeutic agents only in abnormal cells, to document the response to the
therapy, and to identify residual disease.
SUMMARY OF THE INVENTION
The present invention relates to novel therapeutic and diagnostic systems. More
particularly, the present invention is directed to dendrimer based multifunctional
compositions and systems for use in disease diagnosis and therapy (e.g., cancer diagnosis
and therapy). The compositions and systems generally comprise two or more distinct
components for targeting, imaging, sensing, and/or triggering release of a therapeutic or
diagnostic material and monitoring the response to therapy of a cell or tissue (e.g., a
tumor).
For example, the present invention provides a composition comprising a
dendrimer complex, said dendrimer complex comprising first and second dendrimers, the
first dendrimer comprising a first agent and the second dendrimer comprising a second
agent, wherein the first agent is different than the second agent. In preferred
embodiments, the first and said second agents are selected from the group consisting of
therapeutic agents, biological monitoring agents, biological imaging agents, targeting
agents, and agents capable of identifying a specific signature of cellular abnormality. In
some embodiments, the first dendrimer is covalently linked to the second dendrimer. In
certain embodiments, the dendrimer complex includes additional dendrimers. For
example, in some embodiments, the complex comprises a third dendrimer (e.g, a
third-dendrimer covalently linked to the first and second dendrimers). In yet other
embodiments, the dendrimer complex comprises fourth, fifth, or additional dendrimers.
Each of the dendrimers may comprise an agent
In some embodiments, the present invention provides a composition comprising:
a first dendrimer comprising a first agent; and a second dendrimer comprising a second
agent, wherein the first and second dendrimers are complexed (e.g., covalently attached)
with at least one dendrimer (e.g., to each other, to a common third dendrimer, or each
individually to a third and fourth dendrimers respectively), and wherein the first agent is
different than the second agent, and wherein the first and the second agents are selected
from the group consisting of therapeutic agents, biological monitoring agents (i.e., agents
capable of monitoring biological materials or events), biological imaging agents (i.e.,
agents capable of imaging biological materials or events), targeting agents (i.e., agents
capable of targeting a biological material--i. e., specifically interacting with the biological
material), and agents capable of identifying a specific signature of cellular identity (i.e.,
capable of identifying a characteristic of a cell that helps differentiate the cell from other
cell types—e.g., a cellular proteins specific for a particular cellular abnormality). The
present invention is not limited by the nature of the dendrimers. Dendrimers suitable for
use with the present invention include, but are not limited to, polyamidoamine
(PAMAM), polypropylamine (POPAM), polyethylenimine, iptycene, aliphatic poly(ether),
and/or aromatic polyether dendrimers. Each dendrimer of the dendrimer complex may
be of similar or different chemical nature than the other dendrimers (e.g., the first
dendrimer may comprises a PAMAM dendrimer, while the second dendrimer may
comprises a POPAM dendrimer). In some embodiments, the first or second dendrimer
may further comprises an additional agent.
In some embodiments of the present invention, the dendrimer complex may
further comprises one or more additional dendrimers. For example, the composition may
further comprises a third dendrimer, wherein the third-dendrimer is complexed with at
least one other dendrimer. In some embodiments, a third agent is complexed with the
third dendrimer. In some embodiments, the first and second dendrimers are each
complexed to a third dendrimer. In preferred embodiments, the first and second
dendrimers comprise PAMAM dendrimers and the third dendrimer comprises a POPAM
dendrimer. In certain embodiments, the present invention further comprises fourth and/or
fifth dendrimers comprising agents (e.g., third and fourth agents), wherein the fourth
and/or fifth dendrimer is also complexed (e.g., covalently attached) to the third
dendrimer. The present invention is not limited by the number of dendrimers complexed
to one another.
In some embodiments of the present invention, the first agent is a therapeutic
agent and the second agent is a biological monitoring agent In preferred embodiments,
the therapeutic agent includes, but is not limited to, a chemotherapeutic agent, an
anti-oncogenic agent, an anti-vascularizing agent, a anti-microbial or anti-pathogenic
agent, and an expression construct comprising a nucleic acid encoding a therapeutic
protein. In some embodiments, the therapeutic agent is protected with a protecting group
selected from photo-labile, radio-labile, and enzyme-labile protecting groups. In
preferred embodiments, the chemotherapeutic agents include, but are not limited to,
platinum complex, verapamil, podophyllotoxin, carboplatin, procarbazine,
mechlorethamine, cyclophosphamide, camptothecin, ifosfamide, melphalan, chlorambucil,
bisulfan, nitrosurea, adriamycin, dactinomycin, daunorubicin, doxorubicin, bleomycin,
plicomycin, mitomycin, etoposide, tamoxifen, taxol, transplatinum, 5-fluorouracil,
vincristin, vinblastin, and methotrexate. In some embodiments, the anti-oncogenic agent
comprises an antisense nucleic acid. In certain embodiments, the antisense nucleic acid
comprises a sequence complementary to an RNA of an oncogene. In preferred
embodiments, the oncogene includes, but is not limited to, abl, Bcl-2, Bcl-x1, erb, fins,
gsp, hst, jun, myc, neu, raf, ras, ret, src, or trk. In some embodiments, the nucleic acid
encoding a therapeutic protein encodes a factor including, but not limited to, a tumor
suppressor, cytokine, receptor, inducer of apoptosis, or differentiating agent. In preferred
embodiments, the tumor suppressor includes, but is not limited to, BRCA1, BRCA2,
C-CAM, pl6, p21, p53, p73, Rb, and p27. In preferred embodiments, the cytokine
includes, but is not limited to, GMCSF, IL-1, IL-2, IL-3, IL-4, IL-5, EL-6, IL-7, IL-8,
IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, p-interferon, y-interferon, and TNF. In
preferred embodiments, the receptor includes, but is not limited to, CFTR, EGFR,
estrogen receptor, IL-2 receptor, and VEGFR In preferred embodiments, the inducer of
apoptosis includes, but is not limited to, AdE1B, Bad, Bak, Bax, Bid, Bik, Bim, Harakid,
and ICE-CED3 protease. In some embodiments, the therapeutic agent comprises a short-
half life radioisotope.
In some embodiments of the present invention, the biological monitoring agent
comprises an agent that measures an effect of a therapeutic agent (e.g., directly or
indirectly measures a cellular factor or reaction induced by a therapeutic agent), however,
the present invention is not limited by the nature of the biological monitoring agent In
some embodiments, the monitoring agent is capable of measuring the amount of or
detecting apoptosis caused by the therapeutic agent
In some embodiments of the present invention, the imaging agent comprises a
radioactive label including, but not limited to, 14C, 36CI, 57Co, 58Co, 51Cr, ,125l,I3Il, 111ln,
152Eu, 59Fe, 67Ga,32P, 186Re , 35S, 75Se, Tc-99m, and 169Yb, however, the present invention
is not limited by the nature of the imaging agent
In some embodiments of the present invention, the targeting agent includes, but is
not limited to an antibody, receptor ligand, hormone, vitamin, and antigen, however, the
present invention is not limited by the nature of the targeting agent. In some
embodiments, the antibody is specific for a disease specific antigen. In some preferred
embodiments, the disease specific antigen comprises a tumor specific antigen. In some
embodiments, the receptor ligand includes, but is not limited to, a ligand for CFTR,
EGFR, estrogen receptor, FGR2, folate receptor, IL-2 receptor, glycoprotein, and VEGFR.
In some embodiments of fee present invention, the first and second dendrimers
(and third, fourth, . . . ) are attached to one another through linker groups. In some
preferred embodiments, the linker groups comprise nucleic acid linkers. For example, in
some embodiments, the first dendrimer comprises a first nucleic acid linker and the
second dendrimer comprises a second nucleic acid linker, wherein the first nucleic acid
linker is hybridized to the second nucleic acid linker. In some embodiments, a duplex
formed from hybridization of the first linker to the second linker comprises a cleavage
site (e.g., a nuclease recognition site such as a restriction endonuclease site).
The present invention also provides methods for treating a cell with a dendrimer
complex comprising: providing a cell and a composition comprising a dendrimer
complex, and exposing the cell to the dendrimer complex. In some embodiments, the
dendrimer complex comprises a first dendrimer comprising a first agent, and a second
dendrimer comprising a second agent, wherein the first and second dendrimers are
complexed with at least one dendrimer, and wherein the first agent is different than the
second agent, and wherein the first and the second agents are selected from the group
consisting of therapeutic agents, biological monitoring agents, biological imaging agents,
targeting agents, and agents capable of identifying a specific signature of cellular
abnormality; and exposing the cell to the composition. The present invention is not
limited by the nature of the cell type or the exposing step. For example, cells of the
present invention include, but are not limited to, cell residing in vitro (e.g., cell culture
cells) and cells residing in vivo (e.g., cells of a human or animal subject or pathogenic
cells). In preferred embodiments, where the cell resides in a subject (e.g., a human or
animal subject), the subject has a disease (e.g., the cell is a disease cell such as a tumor
cell). In some embodiments, the disease includes, but is not limited to, cancer,
cardiovascular disease, inflammatory disease, and prion-type disease (i.e., diseases
associated with or caused by a prion).
In some embodiments of the present invention, the therapeutic agent is in inactive
form and is rendered active following administration of the composition to the subject.
For example, the agent, upon exposure to light or a change in pH (e.g., due to exposure
to a particular intracellular environment) is altered to assume its active form. In these
embodiments, the agent may be attached to a protective linker (e.g., photo-cleavable,
enzyme-cleavable, pH-cleavable) to make it inactive and become active upon exposure to
the appropriate activating agent (e.g., UV light, a cleavage enzyme, or a change in pH).
In some embodiments of the present invention, the subject has a tumor or is
suspected of having cancer. In certain embodiments the cancer includes, but is not
limited to, lung, breast, melanoma, colon, renal., testicular, ovarian, lung, prostate,
hepatic, germ cancer, epithelial., prostate, head and neck, pancreatic cancer, glioblastoma,
astrocytoma, oligodendroglioma, ependymomas, neurofibrosarcoma, meningia, liver,
spleen, lymph node, small intestine, colon, stomach, thyroid, endometrium, prostate, skin,
esophagus, and bone marrow cancer. In some embodiments, compositions comprising
nanodevices, and any other desired components (e.g., pharmaceutically acceptable
carriers, adjuvants and exipients) are administered to the subject. The present invention
is not limited by the route of administration. Such administration routes include, but are
not limited to, endoscopic, intratracheal., intralesion, percutaneous, intravenous,
subcutaneous, and intratumoral administration.
DESCRIPTION OF THE DRAWINGS
The following drawings form part of the present specification and are included to
further demonstrate certain aspects of the present invention. The invention may be better
understood by reference to one or more of these drawings in combination with the
detailed description of specific embodiments presented herein.
Figure 1 shows several generations of spherical., dendritic polymers, with each
generation increasing the size, molecular weight and number of primary amine groups on
the surface of the polymer.
Figure 2 shows different options for design of dendrimer-based nanodevices.
Figure 3 shows a component structure of nanodevices for breast and colon cancer
in some embodiments of the present invention.
Figures 4A-D show functions of therapeutic nanodevices in some embodiment of
the present invention. Figure 4A shows "targeting and imaging" applications, wherein
the nano-device targets neoplastic cells through a cell-surface moiety and is taken into
the cell through receptor mediated endocytosis. The tumor 00 is imaged through MRI.
Figure 4B shows "sensing cancer signature" applications, wherein red fluorescence is
activated by the presence of the cancer signature (Mucl, Her2, or mutated p53 through
quantum dot-like aggregation or loss of 1 quenching). Figure 4C shows "triggered
release of therapeutic" applications, wherein laser light is targeted to red-emitting cells
and cleaves photo-labile protecting group from drug (e.g., platinum or Taxol releasing it
from dendrimer matrix). Figure 4D shows "monitoring response to therapy" applications,
wherein a drug induces apoptosis in cells, and caspase activity activates green
fluorescence. Apoptotic cancer cells turn orange while residual cancer cells remain red.
Normal cells induced to apoptose (collateral damage) if they fluoresce green.
Figure 5 shows a photograph of an atomic force microscopy (AFM) image of
large (generation 9 MW 800kDA) PAMAM dendritic polymers of the present invention.
There is uniformity in size and shape. Three larger, noncovalently bonded clusters of
dendrimers also are present in the figure.
Figure 6 shows aqueous synthesis of clustered dendrimers in some embodiments
of the present invention.
Figure 7 shows a dendrimer synthesis procedure in some embodiments of the
present invention.
Figure 8 shows a dendrimer synthesis procedure in some embodiments of the
present invention.
Figure 9 shows a graph indicating the toxicity level of certain dendrimers
comprising a therapeutic agent.
Figure 10 shows a representation of a core-shell structure in some embodiments
of the present invention.
Figure 11 shows a representation of a core (Gx-shell structure comprising nucleic
acid linkers in some embodiments of the present invention.
GENERAL DESCRIPTION OF THE INVENTION
The present invention relates to novel therapeutic and diagnostic complexes.
More particularly, the present invention is directed to dendrimer-based multifunctional
compositions and systems for use in disease diagnosis and therapy (e.g., cancer diagnosis
and therapy). The compositions and systems generally comprise two or more separate
components for targeting, imaging, sensing, and/or triggering release of a therapeutic or
diagnostic material and monitoring the response to therapy of a cell or tissue (e.g., a
tumor).
For example, the present invention provides nanodevices comprising two or more
dendrimers, each complexed with one or more components for targeting, imaging,
sensing, and/or triggering release of a therapeutic or diagnostic material and monitoring
the response to therapy of a cell or tissue. In some embodiments of the present
invention, the nanodevice comprises a core dendrimer complexed (e.g., covalently linked)
to other dendrimer subunits containing the above functionalities. The present invention
demonstrates that such compositions are non-toxic and present new methods for treating,
detecting, and monitoring various physiological conditions. For example, in some
embodiments, the nanodevices contain a dendrimer subunit that targets the nanodevice to
particular cells or tissues {e.g., contains binding agents that recognize and are specific
cellular components). In other embodiments, the nanodevices contain a dendrimer
subunit that images a cell, a cellular component, or cellular reactions (e.g., provides a
detectable signal upon exposure to the cell, component, or reaction). In yet other
embodiments, the nanodevices contain a dendrimer subunit that provides a signature
identifying agent such that, directly or indirectly, the presence of a cell or cellular
condition is identified (e.g., identifying a cancer cell through interaction of the signature
identifying agent with a cancer-specific factor). In still further embodiments, the
nanodevices contain a dendrimer subunit that provides a therapeutic or diagnostic agent
for delivery or release into a cell or subject
Thus, the present invention provides a variety of useful therapeutic and diagnostic
compositions for treating and characterizing cells or subjects with various pathologies or
physiological conditions. The nanodevices of the present invention comprises any
number of dendrimer components to give the desired functionality. For example, in
cancer therapy, the present invention provides nanodevices that comprise a core
dendrimer covalently linked to individual dendrimer units comprising signature
identifying agents, imaging agents, therapeutic agents, targeting agents, and monitoring
agents, respectively. For example, for breast cancer {See e.g., Figure 3 showing
complexes for use in breast and colon cancer; and Figure 4 as described above), the core
dendrimer is complexed with a first dendrimer comprising a gadolinium contrast agent
for imaging the tissue by MRI, a second dendrimer comprising a therapeutic agent {e.g.,
Taxol or cisplatin) for treating the cancer, a third dendrimer comprising a ligand for
binding to a folate receptor for targeting the cancer cells, a fourth dendrimer comprising
a fluorogenic component for detecting mutated p53 protein for identifying the cancer
signature, and a fifth dendrimer comprising a fluorogenic marker of apoptosis to monitor
treatment with the therapeutic agent. In some embodiments, the core dendrimer
comprises any of the desired components. In yet other embodiments, two or more of the
functionalities are provided on a single dendrimer.
In preferred embodiments, of the present invention, libraries of individual
dendrimers comprising the above functionalities are created for use in generating any
desired nanodevice complexes. For example, libraries of dendrimers each containing one
of a host of therapeutic agents are created. The same procedure is conducted for target
agents, imaging agents, and the like. Such libraries provide the ability to mix-and-match
components to generate the optimum therapeutic or diagnostic complexes for a desired
application. The nanodevices may be generated rationally, or may be generated
randomly and screened for desired activities. Thus, the present invention provides non-
toxic systems with a wide range of therapeutic and diagnostic uses.
Definitions
To facilitate an understanding of the present invention, a number of terms and
phrases are defined below:
As used herein, the term "dendrimer complex" refers to a complex comprising
two or more dendrimers in physical association with one another (e.g., covalent or non-
covalent attachment to one another). For example, two dendrimers covalently linked to
one another (e.g, directly or through a linking group) provide a dendrimer complex.
As used herein, the term "agent" refers to a composition that possesses a
biologically relevant activity or property. Biologically relevant activities are activities
associated with biological reactions or events or that allow the detection, monitoring, or
characterization biological reactions or events. Biologically relevant activities include,
but are not limited to, therapeutic activities (e.g., the ability to improve biological health
or prevent the continued degeneration associated with an undesired biological condition),
targeting activities (e.g., the ability to bind or associate with a biological molecule or
complex), monitoring activities (e.g., the ability to monitor the progress of a biological
event or to monitor changes in a biological composition), imaging activities (e.g., the
ability to observe or otherwise detect biological compositions or reactions), and signature
identifying activities (e.g., the ability to recognize certain cellular compositions or
conditions and produce a detectable response indicative of the presence of the
composition or condition). The agents of the present invention are not limited to these
particular illustrative examples. Indeed any useful agent may be used including agents
that deliver or destroy biological materials, cosmetic agents, and the like. In preferred
embodiments of the present invention, the agent or agents are associated with at least one
dendrimer (e.g., incorporated into the dendrimer, surface exposed on the dendrimer, etc.).
In some embodiments of the present invention, two or more dendrimers are present in a
composition where any one dendrimer may have an agent that "is different than" an agent
of another dendrimer. "Different than" refers to agents that are distinct from one another
in chemical makeup and/or functionality.
As used herein, the term "nanodevice" refers to small (e.g., invisible to the
unaided human eye) compositions containing or associated with one or more "agents." In
its simplest form, the nanodevice consists of a physical composition (e.g., a dendrimer)
associated with at least one agent that provides biological functionality (e.g., a
therapeutic agent). However, the nanodevice may comprise additional components (e.g.,
additional dendrimers and/or agents). In preferred embodiments of the present invention,
the physical composition of the nanodevice comprises at least one dendrimer and a
biological functionality is provided by at least one agent associated with a dendrimer.
The term "biologically active," as used herein, refers to a protein or other
biologically active molecules (e.g., catalytic RNA) having structural., regulatory, or
biochemical functions of a naturally occurring molecule.
The term "agonist," as used herein, refers to a molecule which, when interacting
with an biologically active molecule, causes a change (e.g., enhancement) in the
biologically active molecule, which modulates the activity of the biologically active
molecule. Agonists may include proteins, nucleic acids, carbohydrates, or any other
molecules which bind or interact with biologically active molecules. For example,
agonists can alter the activity of gene transcription by interacting with RNA polymerase
directly or through a transcription factor.
The terms "antagonist" or "inhibitor," as used herein, refer to a molecule which,
when interacting with a biologically active molecule, blocks or modulates the biological
activity of the biologically active molecule. Antagonists and inhibitors may include
proteins, nucleic acids, carbohydrates, or any other molecules that bind or interact with
biologically active molecules. Inhibitors and antagonists can effect the biology of entire
cells, organs, or organisms (e.g., an inhibitor that slows tumor growth).
The term "modulate," as used herein, refers to a change in the biological activity
of a biologically active molecule. Modulation can be an increase or a decrease in
activity, a change in binding characteristics, or any other change in the biological.,
functional., or immunological properties of biologically active molecules.
The term "gene" refers to a nucleic acid (e.g., DNA) sequence that comprises
coding sequences necessary for the production of a polypeptide or precursor. The
polypeptide can be encoded by a full length coding sequence or by any portion of the
coding sequence so long as the desired activity or functional properties (e.g., enzymatic
activity, ligand binding, signal transduction, etc.) of the full-length or fragment are
retained. The term also encompasses the coding region of a structural gene and the
including sequences located adjacent to the coding region on both the 5' and 3' ends for
a distance of about 1 kb or more on either end such that the gene corresponds to the
length of the full-length mRNA. The sequences that are located 5' of the coding region
and which are present on the mRNA are referred to as 5' non-translated sequences. The
sequences that are located 3' or downstream of the coding region and which are present
on the mRNA are referred to as 3' non-translated sequences. The term "gene"
encompasses both cDNA and genomic forms of a gene. A genomic form or clone of a
gene contains the coding region interrupted with non-coding sequences termed "introns"
or "intervening regions" or "intervening sequences." Introns are segments of a gene
which are transcribed into nuclear RNA (hnRNA); introns may contain regulatory
elements such as enhancers. Introns are removed or "spliced out" from the nuclear or
primary transcript; introns therefore are absent in the messenger RNA (mRNA) transcript.
The mRNA functions during translation to specify the sequence or order of amino acids
in a nascent polypeptide.
As used herein, the terms "nucleic acid molecule encoding," "DNA sequence
encoding," and "DNA encoding" refer to the order or sequence of deoxyribonucleotides
along a strand of deoxyribonucleic acid. The order of these deoxyribonucleotides
determines the order of amino acids along the polypeptide (protein) chain. The DNA
sequence thus codes for the amino acid sequence.
As used herein, the terms "complementary" or "complementarity" are used in
reference to polynucleotides (i.e., a sequence of nucleotides) related by the base-pairing
rules. For example, for the sequence "A-G-T," is complementary to the sequence "T-C-
A." Complementarity may be "partial.," in which only some of the nucleic acids' bases
are matched according to the base pairing rules. Or, there may be "complete" or "total"
complementarity between the nucleic acids. The degree of complementarity between
nucleic acid strands has significant effects on the efficiency and strength of hybridization
between nucleic acid strands. This is of particular importance in amplification reactions,
as well as detection methods that depend upon binding between nucleic acids.
As used herein, the term "hybridization" is used in reference to the pairing of
complementary nucleic acids. Hybridization and the strength of hybridization (i.e., the
strength of the association between the nucleic acids) is impacted by such factors as the
degree of complementary between the nucleic acids, stringency of the conditions
involved, the Tm of the formed hybrid, and the G:C ratio within the nucleic acids.
As used herein, the term "Tm" is used in reference to the "melting temperature."
The melting temperature is the temperature at which a population of double-stranded
nucleic acid molecules becomes half dissociated into single strands. The equation for
calculating the Tm of nucleic acids is well known in the art. As indicated by standard
references, a simple estimate of the Tm value may be calculated by the equation: Tm =
81.5 + 0.41 (% G + C), when a nucleic acid is in aqueous solution at 1 M NaCl (See e.g.,
Anderson and Young, Quantitative Filter Hybridization, in Nucleic Acid Hybridization
[1985]). Other references include more sophisticated computations that take structural as
well as sequence characteristics into account for the calculation of Tm.
As used herein the term "stringency" is used in reference to the conditions of
temperature, ionic strength, and the presence of other compounds such as organic
solvents, under which nucleic acid hybridizations are conducted. Those skilled in the art
will recognize that "stringency" conditions may be altered by varying the parameters just
described either individually or in concert With "high stringency" conditions, nucleic
acid base pairing will occur only between nucleic acid fragments that have a high
frequency of complementary base sequences (e.g., hybridization under "high stringency"
conditions may occur between homologs with about 85-100% identity, preferably about
70-100% identity). With medium stringency conditions, nucleic acid base pairing will
occur between nucleic acids with an intermediate frequency of complementary base
sequences (e.g., hybridization under "medium stringency" conditions may occur between
homologs with about 50-70% identity). Thus, conditions of "weak" or "low" stringency
are often required with nucleic acids that are derived from organisms that are genetically
diverse, as the frequency of complementary sequences is usually less.
"High stringency conditions" when used in reference to nucleic acid hybridization
comprise conditions equivalent to binding or hybridization at 42°C in a solution
consisting of 5X SSPE (43.8 g/1 NaCl, 6.9 g/1 NaH2PO4-H2O and 1.85 g/l EDTA, pH
adjusted to 7.4 with NaOH), 0.5% SDS, 5X Denhardt's reagent and 100 μg/ml denatured
salmon spam DNA followed by washing in a solution comprising 0.1X SSPE, 1.0%
SDS at 42°C when a probe of about 500 nucleotides in length is employed.
"Medium stringency conditions" when used in reference to nucleic acid
hybridization comprise conditions equivalent to binding or hybridization at 42°C in a
solution consisting of 5X SSPE (43.8 g/l NaCl, 6.9 g/l NaH2PO4•H2O and 1.85 g/l
EDTA, pH adjusted to 7.4 with NaOH), 0.5% SDS, 5X Denhardt's reagent and 100
fig/ml denatured salmon sperm DNA followed by washing in a solution comprising 1.0X
SSPE, 1.0% SDS at 42°C when a probe of about 500 nucleotides in length is employed.
"Low stringency conditions" comprise conditions equivalent to binding or
hybridization at 42°C in a solution consisting of 5X SSPE (43.8 g/1 NaCl, 6.9 g/1
NaH2PCyH2O and 1.85 g/1 EDTA, pH adjusted to 7.4 with NaOH), 0.1% SDS, 5X
Denhardt's reagent [50X Denhardt's contains per 500 ml: 5 g Ficoll (Type 400,
Pharamcia), 5 g BSA (Fraction V; Sigma)] and 100 μg/ml denatured salmon sperm DNA
followed by washing in a solution comprising 5X SSPE, 0.1% SDS at 42°C when a
probe of about 500 nucleotides in length is employed.
As used herein, the term "antisense" is used in reference to DNA or RNA
sequences that are complementary to a specific DNA or RNA sequence (e.g., mRNA).
Included within this definition are antisense RNA ("asRNA") molecules involved in gene
regulation by bacteria. Antisense RNA may be produced by any method, including
synthesis by splicing the gene(s) of interest in a reverse orientation to a viral promoter
which permits the synthesis of a coding strand. Once introduced into an embryo, this
transcribed strand combines with natural mRNA produced by the embryo to form
duplexes. These duplexes then block either the further transcription of the mRNA or its
translation. In this manner, mutant phenotypes may be generated. The term "antisense
strand' is used in reference to a nucleic acid strand that is complementary to the "sense"
strand. The designation (-) (i.e., "negative") is sometimes used in reference to the
antisense strand, with the designation (+) sometimes used in reference to the sense (i.e.,
"positive") strand.
The term "antigenic determinant" as used herein refers to that portion of an
antigen that makes contact with a particular antibody (i.e., an epitope). When a protein
or fragment of a protein is used to immunize a host animal., numerous regions of the
protein may induce the production of antibodies which bind specifically to a given region
or three-dimensional structure on the protein; these regions or structures are referred to as
antigenic determinants. An antigenic determinant may compete with the intact antigen
(i.e., the "immunogen" used to elicit the immune response) for binding to an antibody.
The terms "specific binding" or "specifically binding" when used in reference to
the interaction of an antibody and a protein or peptide means that the interaction is
dependent upon the presence of a particular structure (i.e., the antigenic determinant or
epitope) on the protein; in other words the antibody is recognizing and binding to a
specific protein structure rather than to proteins in general. For example, if an antibody
is specific for epitope "A," the presence of a protein containing epitope A (or free,
unlabelled A) in a reaction containing labelled "A" and the antibody will reduce the
amount of labelled A bound to the antibody.
The term "transgene" as used herein refers to a foreign gene that is placed into an
organism by, for example, introducing the foreign gene into newly fertilized eggs or
early embryos. The term "foreign gene" refers to any nucleic acid (e.g., gene sequence)
that is introduced into the genome of an animal by experimental manipulations and may
include gene sequences found in that animal so long as the introduced gene does not
reside in the same location as does the naturally-occurring gene.
As used herein, the term "vector" is used in reference to nucleic acid molecules
that transfer DNA segment(s) from one cell to another. The term "vehicle" is sometimes
used interchangeably with "vector." Vectors are often derived from plasmids,
bacteriophages, or plant or animal viruses.
The term "expression vector" as used herein refers to a recombinant DNA
molecule containing a desired coding sequence and appropriate nucleic acid sequences
necessary for the expression of the operably linked coding sequence in a particular host
organism. Nucleic acid sequences necessary for expression in prokaryotes usually
include a promoter, an operator (optional), and a ribosome binding site, often along with
other sequences. Eukaryotic cells are known to utilize promoters, enhancers, and
termination and polyadenylation signals.
As used herein, the term "gene transfer system" refers to any means of delivering
a composition comprising a nucleic acid sequence to a cell or tissue. For example, gene
transfer systems include, but are not limited to vectors (e.g., retroviral., adenoviral., adeno-
associated viral., and other nucleic acid-based delivery systems), microinjection of naked
nucleic acid, and polymer-based delivery systems (e.g., liposome-based and metallic
particle-based systems). As used herein, the term "viral gene transfer system" refers to
gene transfer systems comprising viral elements (e.g., intact viruses and modified viruses)
to facilitate delivery of the sample to a desired cell or tissue. As used herein, the term
"adenovirus gene transfer system" refers to gene transfer systems comprising intact or
altered viruses belonging to the family Adenoviridae.
The term "transfection" as used herein refers to the introduction of foreign DNA
into eukaryotic cells. Transfection may be accomplished by a variety of means known to
the art including calcium phosphate-DNA co-precipitation, DEAE-dextran-mediated
transfection, polybrene-mediated transfection, electroporation, microinjection, liposome
fusion, lipofection, protoplast fusion, retroviral infection, and biolistics.
As used herein, the term "cell culture" refers to any in vitro culture of cells.
Included within this term are continuous cell lines (e.g., with an immortal phenotype),
primary cell cultures, finite cell lines (e.g., non-transformed cells), and any other cell
population maintained in vitro.
As used herein, the term "in vitro" refers to an artificial environment and to
processes or reactions that occur within an artificial environment. In vitro environments
can consist of, but are not limited to, test tubes and cell culture. The term "in vivo"
refers to the natural environment (e.g., an animal or a cell) and to processes or reaction
that occur within a natural environment.
The term "test compound" refers to any chemical entity, pharmaceutical., drug,
and the like that can be used to treat or prevent a disease, illness, sickness, or disorder of
bodily function. Test compounds comprise both known and potential therapeutic
compounds. A test compound can be determined to be therapeutic by screening using
the screening methods of the present invention. A "known therapeutic compound" refers
to a therapeutic compound that has been shown (e.g., through animal trials or prior
experience with administration to humans) to be effective in such treatment or
prevention.
The term "sample" as used herein is used in its broadest sense and includes
environmental and biological samples. Environmental samples include material from the
environment such as soil and water. Biological samples may be animal., including,
human, fluid (e.g., blood, plasma and serum), solid (e.g., stool), tissue, liquid foods (e.g.,
milk), and solid foods (e.g., vegetables).
As used herein, the terms "photosensitizer," and "photodynamic dye," refer to
materials which undergo transformation to an excited state upon exposure to a light
quantum (hv). Examples of photosensitizers and photodynamic dyes include, but are not
limited to, Photofrin 2, benzoporphyrin, m-tetrahydroxyphenylchlorin, tin etiopurpurin,
copper benzochlorin, and other porphyrins.
DETAILED DESCRIPTION OF THE INVENTION
The present invention provides novel systems and compositions for the treatment
and monitoring of diseases (e.g., cancer). For example, the present invention provides
systems and compositions that target, image, and sense pathophysiological defects,
provide the appropriate therapeutic based on the diseased state, monitor the response to
the delivered therapeutic, and identify residual disease. In preferred embodiments of the
present invention, the compositions are small enough to readily enter a patient's or
subjects cells.
In preferred embodiments, the systems and compositions of the present invention
are used in treatment and monitoring during cancer therapy. However, the systems and
compositions of the present invention find use in the treatment and monitoring of a
variety of disease states or other physiological conditions, and the present invention is not
limited to use with any particular disease state or condition. Other disease states that
find particular use with the present invention include, but are not limited to,
cardiovascular disease, inflammatory disease, and other proliferative disorders.
In preferred embodiments, the present invention provides nanodevices comprising
dendrimer subunits. In preferred embodiments, the nanodevices are limited to a few
hundred nanometers in diameter to facilitate internalization into cells.
Preferred embodiments of the present invention provide a composition comprising
two or more different dendrimer structures, each including at least one functional
component, including, but not limited to, therapeutic agents, biological monitoring
components, biological imaging components, targeting components, and components to
identify the specific signature of cellular abnormalities. These components ultimately
form a therapeutic and/or diagnostic complexes in which each of the different
components is located within a distinct dendrimer carrier. As such, the therapeutic
nanodevice or complex is made up of at least two separate dendrimer carriers being
specifically complexed with or covalently linked to at least one of the other dendrimer
compositions of the complexes.
The following discussion describes individual component parts of the nanodevice
and methods of making and using the same in some embodiments of the present
invention. To illustrate the design and use of the systems and compositions of the
present invention, the discussion focuses on specific embodiments of the use of the
compositions in the treatment and monitoring of breast adenocarcinoma and colon
adenocarcinoma. These specific embodiments are intended only to illustrate certain
preferred embodiments of the present invention and are not intended to limit the scope
thereof. In these embodiments, the nanodevices of the present invention target the
neoplastic cells through cell-surface moieties and are taken up by the tumor cell for
example through receptor mediated endocytosis. The imaging component of the device
allows the tumor to be imaged for example through the use of MRI. In those devices
containing a sensing component, red fluorescence is activated by the presence of the
particular cancer signature (e.g., Mucl, Her2 or mutated p53). This allows a triggered
release of a therapeutic agent contained in the therapeutic component of the nanodevice.
The release is facilitated by the therapeutic component being attached to a labile
protecting group, such as, for example, cisplatin being attached to a photolabile
protecting group that becomes released by laser light directed at those cells emitting the
color of fluorescence activated as mentioned above (e.g., red-emitting) cells. Optionally
the therapeutic device also may have a component to monitor the response of the tumor
to therapy. For example, where the drug induces apoptosis of the cell, the caspase
activity of the cells may be used to activate, a green fluorescence. This allows apoptotic
cells to turn orange, (combination of red and green) while residual cells remain red. Any
normal cells that are induced to undergo apoptosis in collateral damage fluoresce green.
As is clear from the above example, the use of the compositions of the present
invention facilitates non-intrusive sensing, signaling, and intervention for cancer. Since
specific protocols of molecular alterations in cancer cells are identified using this
technique, non-intrusive sensing through the dendritic molecules is achieved and may
then be employed automatically against various tumor phenotypes. If the polymer array
approach is employed, the targeting, sensing, and therapeutic conjugates are interchanged
to address varied tumor types or different pathophysiological alterations. Thus, the array
approach provides common, interchangeable therapeutic platforms that transcend any
single type of tumor or cellular abnormality.
I. Dendrimers
In preferred embodiments of the present invention, the nanodevices comprises
dendrimers. Dendrimeric polymers have been described extensively (See, Tomalia,
Advanced Materials 6:529 [1994]; Angew, Chem, Int. Ed. Engl., 29:138 [1990];
incorporated herein by reference in their entireties). Dendrimers polymers are
synthesized as defined spherical structures typically ranging from 1 to 20 nanometers in
diameter. Several generations of polyamidoamine (B-alanine subunit) dendrimers are
shown in FIG. 1. Molecular weight and the number of terminal groups increase
exponentially as a function of generation (the number of layers) of the polymer.
Different types of dendrimers can be synthesized based on the core structure that initiates
the polymerization process.
The dendrimer core structures dictate several characteristics of the molecule such
as the overall shape, density and surface functionality (Tomalia et al., Chem. Int. Ed.
Engl., 29:5305 [1990]. Spherical dendrimers have ammonia as a trivalent initiator core
or ethylenediamine (EDA) as a tetravalent initiator core. Recently described rod-shaped
dendrimers (Yin et al., J. Am. Chem. Soc, 120:2678 [1998]) use polyemyleneimine
linear cores of varying lengths; the longer the core, the longer the rod. Dendritic
macromolecules are available commercially in kilogram quantities and are produced
under current good manufacturing processes (GMP) for biotechnology applications.
Dendrimers may be characterized by a number of techniques including, but not
limited to, electrospray-ionization mass spectroscopy, 13C nuclear magnetic resonance
spectroscopy, high performance liquid chromatography, size exclusion chromatography
with multi-angle laser light scattering, capillary electrophoresis and gel electrophoresis.
These tests assure the uniformity of the polymer population and are important for
monitoring quality control of dendrimer manufacture for GMP applications and in vivo
usage. Extensive studies have been completed with dendrimers and show no evidence of
toxicity when administered intravenously in in vivo studies (Roberts et al., J. Biomed.
Mat Res., 30:53 [1996] and Bourne et al., J. Magn. Reson. Imag., 6:305 [1996]).
Numerous U.S. Patents describe methods and compositions for producing
dendrimers. Examples of some of these patents are given below in order to provide a
description of some dendrimer compositions that may be useful in the present invention,
however it should be understood that these are merely illustrative examples and
numerous other similar dendrimer compositions could be used in the present invention.
U.S. Patent 4,507,466, U.S. Patent 4,558,120, U.S. Patent 4,568,737, and U.S.
Patent 4,587,329 each describe methods of making dense star polymers with terminal
densities greater than conventional star polymers. These polymers have greater/more
uniform reactivity than conventional star polymers, i.e. 3rd generation dense star
polymers. These patents further describe the nature of the amidoamine dendrimers and
the 3-dimensional molecular diameter of the dendrimers.
U.S. Patent 4,631,337 describes hydrolytically stable polymers. U.S. Patent
4,694,064 describes rod-shaped dendrimers. U.S. Patent 4,713,975 describes dense star
polymers and their use to characterize surfaces of viruses, bacteria and proteins including
enzymes. Bridged dense star polymers are described in U.S. Patent 4,737,550. U.S.
Patent 4,857,599 and U.S. Patent 4,871,779 describe dense star polymers on immobilized
cores useful as ion-exchange resins, chelation resins and methods of making such
polymers.
U.S. Patent 5,338,532 is directed to starburst conjugates of dendrimer(s) in
association with at least one unit of carried agricultural., pharmaceutical or other material.
This patent describes the use of dendrimers to provide means of delivery of high
concentrations of carried materials per unit polymer, controlled delivery, targeted delivery
and/or multiple species such as e.g., drugs antibiotics, general and specific toxins, metal
ions, radionuclides, signal generators, antibodies, interleukins, hormones, interferons,
viruses, viral fragments, pesticides, and antimicrobials.
Other useful dendrimer type compositions are described in U.S. Patent 5,387,617,
U.S. Patent 5,393,797, and U.S. Patent 5,393,795 in which dense star polymers are
modified by capping with a hydrophobic group capable of providing a hydrophobic outer
shell. U.S. Patent 5,527,524 discloses the use of amino terminated dendrimers in
antibody conjugates.
The use of dendrimers as metal ion carriers is described in U.S. Patent 5,560,929.
U.S. Patent 5,773,527 discloses non-crosslinked polybranched polymers having a
comb-burst configuration and methods of making the same. U.S. Patent 5,631,329
describes a process to produce polybranched polymer of high molecular weight by
forming a first set of branched polymers protected from branching; grafting to a core;
deprotecting first set branched polymer, then forming a second set of branched polymers
protected from branching and grafting to the core having the first set of branched
polymers, etc.
U.S. Patent 5,902,863 describes dendrimer networks containing lipophilic
organosilicone and hydrophilic polyanicloamine nanscopic domains. The networks are
prepared from copolydendrimer precursors having PAMAM (hydrophilic) or
polyproyleneimine interiors and organosilicon; outer layers. These dendrimers have a
controllable size, shape and spatial distribution. They are hydrophobic dendrimers with
an organosilicon outer layer that can be used for specialty membrane, protective coating,
composites containing organic organometallic or inorganic additives, skin patch delivery,
absorbants, chromatography personal care products and agricultural products.
U.S. Patent 5,795,582 describes the use of dendrimers as adjutants for influenza
antigen. Use of the dendrimers produces antibody titer levels with reduced antigen dose.
U.S. Patent 5,898,005 and U.S. Patent 5,861,319 describe specific immunobinding assays
for determining concentration of an analyte. U.S. Patent 5,661,025 provides details of a
self-assembling polynucleotide delivery system comprising dendrimer polycation to aid in
delivery of nucleotides to target site. This patent provides methods of introducing a
polynucleotide into a eukaryotic cell in vitro comprising contacting the cell with a
composition comprising a polynucleotide and a dendrimer polycation non-covalently
coupled to the polynucleotide.
Dendrimer-antibody conjugates for use in in vitro diagnostic applications has
previously been demonstrated (Singh et al., Clin. Chem., 40:1845 [1994]), for the
production of dendrimer-chelant-antibody constructs, and for the development of
boronated dendrimer-antibody conjugates (for neutron capture therapy); each of these
latter compounds may be used as a cancer therapeutic (Wu et al., Bioorg. Med. Chem.
Lett, 4:449 [1994]; Wiener et al., Magn. Reson. Med. 31:1 [1994]; Barth et al.,
Bioconjugate Chem. 5:58 [1994]; and Barth et al).
Some of these conjugates have also been employed in the magnetic resonance
imaging of tumors (Wu et al., [1994] and Wiener et al., [1994], supra). Results from
this work have documented that, when administered in vivo, antibodies can direct
dendrimer-associated therapeutic agents to antigen-bearing tumors. Dendrimers also have
been shown to specifically enter cells and carry either chemotherapeutic agents or genetic
therapeutics. In particular, studies show that cisplatin encapsulated in dendrimer
polymers has increased efficacy and is less toxic than cisplatin delivered by other means
(Duncan and Malik, Control Rel. Bioact. Mater. 23:105 [1996]).
Dendrimers have also been conjugated to fluorochromes or molecular beacons and
shown to enter cells. They can then be detected within the cell in a manner compatible
with sensing apparatus for evaluation of physiologic changes within cells (Baker et ai,
Anal. Chem. 69:990 [1997]). Finally, dendrimers have been constructed as differentiated
block copolymers where the outer portions of the molecule may be digested with either
enzyme or light-induced catalysis (Urdea and Horn, Science 261:534 [1993]). This
would allow the controlled degradation of the polymer to release therapeutics at the
disease site and could provide a mechanism for an external trigger to release the
therapeutic agents.
While single dendrimers have been shown to contain these particular functions, to
date, there has been no demonstration of a device that encompasses more than one of
these modalities in a specific configuration. The present invention provides such
nanodevices, wherein two or more dendrimers, each with a specific functionality are
combined into a single complex. For example, preferred complexes of the present
invention are constructed from individual dendrimer modules around a core dendrimer.
This provides a core-shell dendrimer or a cluster molecule as shown in Figure 2. Prior
to the constuction of the multi-dendrimer complex, separate conjugates for each of the
different activities, e.g., one dendrimer conjugate for sensing, one for targeting and
another for therapeutic carrier are produced. These different dendrimer modules are then
clustered together and covalently linked in a manner that yields a single therapeutic
device or complex.
In this approach, one dendrimer acts as a core around which other shell-type
dendrimers are covalently attached. In a preferred embodiment, the core molecule is an
arnine-terminated dendrimer. The shell reagent dendrimers possess carboxylic acid/ester
groups that allow covalent attachment by amide formation to the core. A highly
concentrated mix of ammo-terminated dendrimers with different functional groups of the
same or higher generation are then added to a core dendrimer. A cluster then forms by
amide formation between the terminal amine groups of the core and the free terminal
carboxylic acid groups of the functional outer dendrimers. A limited number of bonds
can form between the core dendrimer and each outer-layer dendrimer because of
sterically induced stoichiometries. In some embodiments, a molar excess of the
outer-layer dendrimer is used to bias the reaction so that each outer core dendrimer reacts
only with a single core molecule.
II. Therapeutic Agents
A wide range of therapeutic agents find use with the present invention. Any
therapeutic agent that can be associated with a dendrimer may be delivered using the
methods, systems, and compositions of the present invention. To illustrate delivery of
therapeutic agents, the following discussion focuses mainly on the delivery of cisplatin
and taxol for the treatment of cancer. Also discussed are various photodynamic therapy
compounds, and various antimicrobial compounds.
i. Cisplatin and Taxol
Cisplatin and Taxol have a well-defined action of inducing apoptosis in tumor
cells {See e.g., Lanni et al., Proc. Natl. Acad. Sci., 94:9679 [1997]; Tortora et al., Cancer
Research 57:5107 [1997]; and Zaffaroni et al., Brit. J. Cancer 77:1378 [1998]).
However, treatment with these and other chemotherapeutic agents is difficult to
accomplish without incurring significant toxicity. The agents currently in use are
generally poorly water soluble, quite toxic, and given at doses that affect normal cells as
wells as diseased cells. For example, paclitaxel (Taxol), one of the most promising
anticancer compounds discovered, is poorly soluble in water. Paclitaxel has shown
excellent antitumor activity in a wide variety of tumor models such as the B16
melanoma, L1210 leukemias, MX-1 mammary tumors, and CS-1 colon tumor xenografts.
However, the poor aqueous solubility of paclitaxel presents a problem for human
administration. Accordingly, currently used paclitaxel formulations require a cremaphor
to solubilize the drug. The human clinical dose range is 200-500 mg. This dose is
dissolved in a 1:1 solution of ethanol:cremaphor and diluted to one liter of fluid given
intravenously. The cremaphor currently used is polyemoxylated castor oil. It is given by
infusion by dissolving in the cremaphor mixture and diluting with large volumes of an
aqueous vehicle. Direct administration (e.g., subcutaneous) results in local toxicity and
low levels of activity. Thus, there is a need for more efficient and effective delivery
systems for these chemomerapeutic agents.
The present invention overcomes these problems by providing methods and
compositions for specific drug delivery. The present invention also provides the ability
to administer combinations of agents {e.g., two or more different merapeutic agents) to
produce an additive effect. The use of multiple agent may be used to counter disease
resistance to any single agent For example, resistance of some cancers to single drugs
(taxol) has been reported (Yu et al., Molecular Cell. 2:581 [1998]). Experiments
conducted during the development of the present invention have demonstrated that
cisplatin, conjugated to dendrimers, is even able to efficiently kill cancer cells that are
resistant to cisplatin (See, Example 4). Although an understanding of the mechanism is
not necessary to practice the present invention and the present invention is not so limited,
it is contemplated that the dendrimer conjugates prevent multidrug resistance channels
from pumping the cisplatin out of the cell.
The present invention also provides the opportunity to monitor therapeutic success
following delivery of cisplatin and/or Taxol to a subject. For example, measuring the
ability of these drugs to induce apoptosis in vitro is reported to be a marker for in vivo
efficacy (Gibb, Gynecologic Oncology 65:13 [1997]). Therefore, in addition to the
targeted delivery of either one or both of these drugs to provide effective anti-tumor
therapy and reduce toxicity, the effectiveness of the therapeutic can be gauged by
techniques of the present invention that monitor the induction of apoptosis. Importantly,
both therapeutics are active against a wide-range of tumor types including, but not
limited to, breast cancer and colon cancer (Akutsu et al., Eur. J. Cancer 31A:2341
[1995]).
Although the above discussion describes two specific agents, any pharmaceutical
that is routinely used in a cancer therapy context finds use in the present invention. In
treating cancer according to the invention, the therapeutic component of the nanodevice
may comprise compounds including, but not limited to, adriamycin, 5-fluorouracil,
etoposide, camptothecin, actinomycin-D, mitomycin C, or more preferably, cisplatin.
The agent may be prepared and used as a combined therapeutic composition, or kit, by
combining it with the immunotherapeutic agent, as described herein.
In some embodiments of the present invention, the dendrimer systems further
comprise one or more agents that directly cross-link nucleic acids (e.g., DNA) to
facilitate DNA damage leading to a synergistic, antineoplastic agents of the present
invention. Agents such as cisplatin, and other DNA alkylating agents may be used.
Cisplatin has been widely used to treat cancer, with efficacious doses used in clinical
applications of 20 mg/M2 for 5 days every three weeks for a total of three courses. The
nanodevice may be delivered via any suitable method, including, but not limited to,
injection intravenously, subcutaneously, intratumorally, intraperitoneally, or topically
(e.g., to mucosal surfaces).
Agents that damage DNA also include compounds that interfere with DNA
replication, mitosis and chromosomal segregation. Such chemotherapeutic compounds
include adriamycin, also known as doxorubicin, etoposide, verapamil, podophyllotoxin,
and the like. Widely used in a clinical setting for the treatment of neoplasms, these
compounds are administered through bolus injections intravenously at doses ranging from
25-75 Mg/M2 at 21 day intervals for adriamycin, to 35-50 Mg/M2 for etoposide
intravenously or double the intravenous dose orally.
Agents that disrupt the synthesis and fidelity of nucleic acid precursors and
subunits also lead to DNA damage and find use as chemotherapeutic agents in the
present invention. A number of nucleic acid precursors have been developed.
Particularly useful are agents that have undergone extensive testing and are readily
available. As such, agents such as 5-fluorouracii (5-FU) are preferentially used by
neoplastic tissue, making this agent particularly useful for targeting to neoplastic cells.
The doses delivered may range from 3 to 15 mg/kg/day, although other doses may vary
considerably according to various factors including stage of disease, amenability of the
cells to the therapy, amount of resistance to the agents and the like.
The anti-cancer therapeutic agents that find use in the present invention are those
that are amenable to incorporation into dendrimeric structures or are otherwise associated
with dendrimer structures such that they can be delivered into a subject, tissue, or cell
without loss of fidelity of its anticancer effect. For a more detailed description of cancer
therapeutic agents such as a platinum complex, verapamil, podophyllotoxin, carboplatin,
procarbazine, mechoremamine, cyclophosphamide, camptothecin, ifosfamide, melphalan,
chlorambucil, bisulfan, nitrosurea, adriamycin, dactinomycin, daunorubicin, doxorubicin,
bleomycin, plicomycin, mitomycin, etoposide (VP16), tamoxifen, taxol, transplatinum,
5-fluorouracil, vincristin, vinblastin and methotrexate and other similar anti-cancer
agents, those of skill in the art are referred to any number of instructive manuals
including, but not limited to, the Physician's Desk reference and to Goodman and
Gilman's "Pharmaceutical Basis of Therapeutics" ninth edition, Eds. Hardman et al.,
1996.
In preferred embodiments, the drugs are preferably attached to the nanodevice
with photocleavable linkers. For example, several heterobifunctional., photocleavable
linkers that find use with the present invention are described by Ottl et al (Ottl et al.,
Bioconjugate Chem., 9:143 [1998]). These linkers can be either water or organic
soluble. They contain an activated ester that can react with amines or alcohols and an
epoxide that can react with a thiol group. In between the two groups is a 3,4-dimethoxy-
6-nitrophenyl photoisomerization group, which, when exposed to near-ultraviolet light
(365 nm), releases the amine or alcohol in intact form. Thus, the therapeutic agent,
when linked to the compositions of the present invention using such linkers, may be
released in biologically active or activatable form through exposure of the target area to
near-ultraviolet light.
In an exemplary embodiment, the alcohol group of taxol is reacted with the
activated ester of the organic-soluble linker. This product in turn is reacted with the
partially-thiolated surface of appropriate dendrimers (the primary amines of the
dendrimers can be partially converted to thiol-containing groups by reaction with a
sub-stoichiometric amount of 2-iminothiolano). In the case of cisplatin, the amino
groups of the drug are reacted with the water-soluble form of the linker. If the amino
groups are not reactive enough, a primary ammo-containing active analog of cisplatin,
such as Pt(II) sulfadiazine dichloride (Pasani et al., Inorg. Chim. Acta 80:99 [1983] and
Abel et al., Eur. J. Cancer 9:4 [1973]) can be used. Thus conjugated, the drug is
inactive and will not harm normal cells. When the conjugate is localized within tumor
cells, it is exposed to laser light of the appropriate near-UV wavelength, causing the
active drug to be released into the cell.
Similarly, in other embodiments of the present invention, the amino groups of
cisplatin (or an analog thereof) is linked with a very hydrophobic photocleavable
protecting group, such as the 2-nitrobenzyloxycarbonyl group (Pillai, V.N.R. Synuiesis:
1-26 [1980]). With this hydrophobic group attached, the drug is loaded into and very
preferentially retained by the hydrophobic cavities within the PAMAM dendrimer (See
e.g., Esfand et al., Pharm. Sci., 2:157 [1996]), insulated from the aqueous environment
When exposed to near-UV light (about 365 nm), the hydrophobic group is cleaved,
leaving the intact drug. Since the drug itself is hydrophilic, it diffuses out of the
dendrimer and into the tumor cell, where it initiates apoptosis.
An alternative to photocleavable linkers are enzyme cleavable linkers. A number
of photocleavable linkers have been demonstrated as effective anti-tumor conjugates and
can be prepared by attaching cancer therapeutics, such as doxorubicin, to water-soluble
polymers with appropriate short peptide linkers (See e.g., Vasey et ai, Clin. Cancer Res.,
5:83 [1999]). The linkers are stable outside of the cell, but are cleaved by thiolproteases
once within the cell In a preferred embodiment, the conjugate PK1 is used As an
alternative to the photocleavable linker strategy, enzyme-degradable linkers, such as
Gly-Phe-Leu-Gly may be used.
The present invention is not limited by the nature of the therapeutic technique.
For example, other conjugates that find use with the present invention include, but are
not limited to, using conjugated boron dusters for BNCT (Capala et al., Bioconjugate
Chem., 7:7 [1996]), the use of radioisotopes, and conjugation of toxins such as ricin to
the nanodevice.
ii. Photodynamic Therapy
Photodynamic therapeutic agents may also be used as therapeteutic agents in the
present invention. In some embodiments, the dendrimeric compositions of the present
invention containing photodynamic compounds are illuminated, resulting in the
production of singlet oxygen and free radicals that diffuse out of the fiberless radiative
effector to act on the biological target (e.g., tumor cells or bacterial cells). Some
preferred photodynamic compounds include, but are not limited to, those that can
participate in a type II photochemical reaction:

where PS = photosenstizer, PS*(1) = excited singlet state of PS, PS*(3) = excited triplet
state of PS, hv = light quantum, *02 = excited singlet state of oxygen, and T =
biological target. Other photodynamic compounds useful in the present invention include
those that cause cytotoxity by a different mechanism than singlet oxygen production
(e.g., copper benzochlorin, Selman, et al., Photochem. Photobiol., 57:681-85 [1993],
incorporated herein by reference). Examples of photodynamic compounds that find use
in the present invention include, but are not limited to Photofrin 2, phtalocyanins (See
e.g., Brasseur et al., Photochem. Photobiol., 47:705-11 [1988]), benzoporphyrin,
tetrahydroxyphenylporphyrins, naphtalocyanines (See e.g., Firey and Rodgers,
Photochem. Photobiol., 45:535-38 [1987]), sapphyrins (Sessler et al., Proc. SPEE,
1426:318-29 [1991]), porphinones (Chang et al., Proc. SPIE, 1203:281-86 [1990]), tin
etiopurpurin, ether substituted porphyrins (Pandey et al., Photochem. Photobiol., 53:65-72
[1991]), and cationic dyes such as the phenoxazines (See e.g., Cincotta et al., SPIE Proa,
1203:202-10 [1590]).
In other embodiments, toxic agents that directly produce free radicals (i.e., do not
produce singlet oxygen) are incorporated into the fiberless radiative effectors during
polymerization. This approach allows for larger and longer lived fiberless radiative
effectors and will not be limited by local oxygen supplies. Such toxic agents include, but
are not limited to 2-methyl-4-nitro-quinoline-N-oxide (Aldrich) and 4,4-dinitro-(2,2)
bipyridinyl-N,N dioxide (Aldrich), which produce hydroxyl radicals when illuminated
with 360-400 nm light (Botchway et al., Photochem. PhotobioL 67(7):635-40 [1998]);
malachite green and isofuran blue (Molecular Probes), which produce hydroxyl radicals
upon stimulation with about 630 nm light (Jay et al., PNAS 91:2659 [1994]; Haugland,
Handbook of Fluorescent Probes and Research Chemicals, 6th ed., Molecular Probes,
Eugene, OR [1994]); potassium nitrosylpentachloromthenate (Molecular Probes) (abs =
516 nm), Roussin's black salt and Roussin's red salt (abs 313-546 nm), serve as sources
of NO which is toxic to cells (Murphy et al., Neuropharm. 33:1375-85 [1994]; Bourassa
et al., JACS 119:2853-60 [1997]); and other photolytic nitric oxide and hydroxyl donors
(De Leo and Ford, JACS 121:1980-81 [1999]).
iii. Antimicrobial Therapeutic Agents
Antimicrobial therapeutic agents may also be used as therapeteutic agents in the
present invention. Any agent that can kill, inhibit, or otherwise attenuate the function of
microbial organisms may be used, as well as any agent contemplated to have such
activities. Antimicrobial agents include, but are not limited to, natural and synthetic
antibiotics, antibodies, inhibitory proteins, antisense nucleic acids, membrane disruptive
agents and the like, used alone or in combination. Indeed, any type of antibiotic may be
used including, but not limited to, anti-bacterial agents, anti-viral agents, anti-fungal
agents, and the like.
III. Signature Identifying Agents
In certain embodiments, the nano-devices of the present invention contain one or
more signature identifying agents that are activated by, or are able to interact with, a
signature component ("signature"). In preferred embodiments, the signature identifying
agent is an antibody, preferably a monoclonal antibody, that specifically binds the
signature {e.g., cell surface molecule specific to a cell to be targeted).
In some embodiments of the present invention, tumor cells are identified. Tumor
cells have a wide variety of signatures, including the defined expression of
cancer-specific antigens such as Mucl, HER-2 and mutated p53 in breast cancer. These
act as specific signatures for the cancer, being present in 30% (HER-2) to 70% (mutated
p53) of breast cancers. In a preferred embodiment, a nanodevice of the present invention
comprises a monoclonal antibody mat specifically binds to a mutated version of p53 that
is present in breast cancer.
In some embodiments of the present invention, cancer cells expressing
susceptibility genes are identified. For example, in some embodiments, there are two
breast cancer susceptibility genes that are used as specific signatures for breast cancer:
BRCA1 on chromosome 17 and BRCA2 on chromosome 13. When an individual carries
a mutation in either BRCA1 or BRCA2, they are at an increased risk of being diagnosed
with breast or ovarian cancer at some point in their lives. These genes participate in
repairing radiation-induced breaks in double-stranded DNA. It is thought that mutations
in BRCA1 or BRCA2 might disable this mechanism, leading to more errors in DNA
replication and ultimately to cancerous growth.
In addition, the expression of a number of different cell surface receptors find use
as targets for the binding and uptake of the nano-device. Such receptors include, but are
not limited to, EGF receptor, folate receptor, FGR receptor 2, and the like.
In some embodiments of the present invention, changes in gene expression
associated with chromosomal abborations are the signature component. For example,
Burkitt lymphoma results from chromosome translocations that involve the Myc gene. A
chromosome translocation means that a chromosome is broken, which allows it to
associate with parts of other chromosomes. The classic chromosome translocation in
Burkitt lymophoma involves chromosome 8, the site of the Myc gene. This changes the
pattern of Myc expression, thereby disrupting its usual function in controlling cell growth
and proliferation.
In other embodiments, gene expression associated with colon cancer are identified
as the signature component. Two key genes are known to be involved in colon cancer:
MSH2 on chromosome 2 and MLH1 on chromosome 3. Normally, the protein products
-of these genes help to repair mistakes made in DNA replication. If the MSH2 and
MLH1 proteins are mutated, the mistakes in replication remain unrepaired, leading to
damaged DNA and colon cancer. MEN1 gene, involved in multiple endocrine neoplasia,
has been known for several years to be found on chromosome 11, was more finely
mapped in 1997, and serves as a signature for such cancers. In preferred embodiments
of the present invention, an antibody specific for the altered protein or for the expressed
gene to be detected is complexed with nanodevices of the present invention.
In yet another embodiment, adenocarcinoma of the colon has defined expression
of CEA and mutated p53, both well-documented rumor signatures. The mutations of p53
in some of these cell lines are similar to that observed in some of the breast cancer cells
and allows for the sharing of a p53 sensing component between the two nanodevices for
each of these cancers (i.e., in assembling the nanodevice, dendrimers comprising the
same signature identifying agent may be used for each cancer type). Both colon and
breast cancer cells may be reliably studied using cell lines to produce tumors in nude
mice, allowing for optimization and characterization in animals.
From the discussion above it is clear that there are many different tumor
signatures that find use with the present invention, some of which are specific to a
particular type of cancer and others which are promiscuous in their origin. The present
invention is not limited to any particular tumor signature or any other disease-specific
signature. For example, tumor suppressors that find use as signatures in the present
invention include, but are not limited to, p53, Mucl, CEA, pl6, p21, p27, CCAM, RB,
APC, DCC, NF-1, NF-2, WT-1, MEN-1, MEN-II, p73, VHL, FCC and MCC.
IV. Biological Imaging Component
In some embodiments of the present invention, the nanodevice comprises at least
one dendrimer-based nanoscopic building block that can be readily imaged. The present
invention is not limited by the nature of the imaging component used. In some
embodiments of the present invention, imaging modules comprise surface modifications
of quantum dots (See e.g., Chan and Nie, Science 281:2016 [1998]) such as zinc
sulfide-capped cadmium selenide coupled to biomolecules (Sooklal., Adv. Mater., 10:1083
[1998]).
However, in preferred embodiments, the imaging module comprises dendrimers
produced according to the "nanocomposite" concept (Balogh et al., Proc. of ACS PMSE
77:118 [1997] and Balogh and Tomalia, J. Am. Che. Soc, 120:7355 [1998]). In these
embodiments, dendrimers are produced by reactive encapsulation, where a reactant is
preorganized by the dendrimer template and is then subsequently immobilized in/on the
polymer molecule by a second reactant. Size, shape, size distribution and surface
functionality of these nanoparticles are determined and controlled by the dendritic
macromolecules. These materials have the solubility and compatibility of the host and
have the optical or physiological properties of the guest molecule (i.e., the molecule that
permits imaging). While the dendrimer host may vary according to the medium, it is
possible to load the dendrimer hosts with different compounds and at various guest
concentration levels. Complexes and composites may involve the use of a variety of
metals or other inorganic materials. The high electron density of these materials
considerably simplifies the imaging by electron microscopy and related scattering
techniques. In addition, properties of inorganic atoms introduce new and measurable
properties for imaging in either the presence or absence of interfering biological
materials. In some embodiments of the present invention, encapsulation of gold, silver,
cobalt, iron atoms/molecules and/or organic dye molecules such as fluorescein are
encapsulated into dendrimers for use as nanoscopi composite labels/tracers, although any
material that facilitates imaging or detection may be employed.
In some embodiments of the present invention, imaging is based on the passive or
active observation of local differences in density of selected physical properties of the
investigated complex matter. These differences may be due to a different shape (e.g.,
mass density detected by atomic force microscopy), altered composition (e.g.,
radiopaques detected by X-ray), distinct light emission (e.g., fluorochromes detected by
spectrophotometry), different diffraction (e.g., electron-beam detected by TEM),
contrasted absorption (e.g., light detected by optical methods), or special radiation
emission (e.g., isotope methods), etc. Thus, quality and sensitivity of imaging depend on
the property observed and on the technique used. The imaging techniques for cancerous
cells have to provide sufficient levels of sensitivity to observe small, local concentrations
of selected cells. The earliest identification of cancer signatures requires high selectivity
(i.e., highly specific recognition provided by appropriate targeting) and the highest
possible sensitivity.
A. Magnetic Resonance Imaging
Once the targeted nanodevice has attached to (or been internalized into) tumor
cells, one or more modules on the device serve to image its location. Dendrimers have
already been employed as biomedical imaging agents, perhaps most notably for magnetic
resonance imaging (MRI) contrast enhancement agents (See e.g., Wiener et al., Mag.
Reson. Med. 31:1 [1994]; an example using PAMAM dendrimers). These agents are
typically constructed by conjugating chelated paramagnetic ions, such as
Gd(III)-diemylenetriaminepentaacetic acid (Gd(IIT)-DTPA), to water-soluble dendrimers.
Other paramagnetic ions that may be useful in this context of the include, but are not
limited to, gadolinium, manganese, copper, chromium, iron, cobalt, erbium, nickel,
europium, technetium, indium, samarium, dysprosium, ruthenium, ytterbium, yttrium, and
holmium ions and combinations thereof. In some embodiments of the present invention,
the dendrimer is also conjugated to a targeting group, such as epidermal growth factor
(EGF), to make the conjugate specifically bind to the desired cell type (e.g., in the case
of EGF, EGFR-expressing tumor cells). In a preferred embodiment of the present
invention, DTPA is attached to dendrimers via the isothiocyanate of DTPA as described
by Wiener (Wiener et al., Mag. Reson. Med. 31:1 [1994]).
Dendrimeric MRI agents are particularly effective due to the polyvalency, size
and architecture of dendrimers, which results in molecules with large proton relaxation
enhancements, high molecular relaxivity, and a high effective concentration of
paramagnetic ions at the target site. Dendrimeric gadolinium contrast agents have even
been used to differentiate between benign and malignant breast tumors using dynamic
MRI, based on how the vasculature for the latter type of tumor images more densely
(Adam et al., Ivest. Rad. 31:26 [1996]). Thus, MRI provides a particularly useful
imaging system of the present invention.
B. Microscopic Imaging
Static structural microscopic imaging of cancerous cells and tissues has
traditionally been performed outside of the patient. Classical histology of tissue biopsies
provides a fine illustrative example, and has proven a powerful adjunct to cancer
diagnosis and treatment. After removal., a specimen is sliced thin (e.g., less than 40
microns), stained, fixed, and examined by a pathologist. If images are obtained, they are
most often 2-D transmission bright-field projection images. Specialized dyes are
employed to provide selective contrast, which is almost absent from the unstained tissue,
and to also provide for the identification of aberrant cellular constituents. Quantifying
sub-cellular structural features by using computer-assisted analysis, such as in nuclear
ploidy determination, is often confounded by the loss of histologic context owing to the
thinness of the specimen and the overall lack of 3-D information. Despite the limitations
of the static imaging approach, it has been invaluable to allow for the identification of
neoplasia in biopsied tissue. Furthermore, its use is often the crucial factor in the
decision to perform invasive and risky combinations of chemotherapy, surgical
procedures, and radiation treatments, which are often accompanied by severe collateral
tissue damage, complications, and even patient death.
The nanodevices of the present invention allow functional microscopic imaging of
tumors and provide improved methods for imaging. The methods find use in vivo, in
vitro, and ex vivo. For example, in one embodiment of the present invention, dendrimers
of the present invention are designed to emit light or other detectable signals upon
exposure to light. Although the labeled dendrimers may be physically smaller than the
optical resolution limit of the microscopy technique, they become self-luminous objects
when excited and are readily observable and measurable using optical techniques. In
some embodiments of the present invention, sensing fluorescent biosensors in a
microscope involves the use of tunable excitation and emission filters and
multiwavelength sources (Farkas et al., SPEI 2678:200 [1997]). In embodiments where
the imaging agents are present in deeper tissue, longer wavelengths in the Near-infrared
(NIR) are used (See e.g., Lester et al., Cell Mol. Biol. 44:29 [1998]). Dendrimeric
biosensing in the Near-IR has been demonstrated with dendrimeric biosensing
antenna-like architectures (Shortreed et al., J. Phys. Chem., 101:6318 [1997]).
Biosensors that find use with the present invention include, but are not limited to,
fluorescent dyes and molecular beacons.
In some embodiments of the present invention, in vivo imaging is accomplished
using functional imaging techniques. Functional imaging is a complementary and
potentially more powerful techniques as compared to static structural imaging.
Functional imaging is best known for its application at the macroscopic scale, with
examples including functional Magnetic Resonance Imaging (fMRI) and Positron
Emission Tomography (PET). However, functional microscopic imaging may also be
conducted and find use in in vivo and ex vivo analysis of living tissue. Functional
microscopic imaging is an efficient combination of 3-D imaging, 3-D spatial
multispectral volumetric assignment, and temporal sampling: in short a type of 3-D
spectral microscopic movie loop. Interestingly, cells and tissues autofluoresce. When
excited by several wavelengths, providing much of the basic 3-D structure needed to
characterize several cellular components (e.g., the nucleus) without specific labeling.
Oblique light illumination is also useful to collect structural information and is used
routinely. As opposed to structural spectral microimaging, functional spectral
microimaging may be used with biosensors, which act to localize physiologic signals
within the cell or tissue. For example, in some embodiments of the present invention,
biosensor-comprising dendrimers of the present invention are used to image upregulated
receptor families such as the folate or EGF classes. In such embodiments, functional
biosensing therefore involves the detection of physiological abnormalities relevant to
carcinogenesis or malignancy, even at early stages. A number of physiological
conditions may be imaged using the compositions and methods of the present invention
including, but not limited to, detection of nanoscopic dendrimeric biosensors for pH,
oxygen concentration, Ca2+ concentration, and other physiologically relevant analytes.
V. Biological Monitoring Component
The biological monitoring or sensing component of the nanodevice of the present
invention is one which that can monitor the particular response in the tumor cell induced
by an agent (e.g., a therapeutic agent provided by the therapeutic component of the
nanodevice). While the present invention is not limited to any particular monitoring
system, the invention is illustrated by methods and compositions for monitoring cancer
treatments. In preferred embodiments of the present invention, the agent induces
apoptosis in cells and monitoring involves the detection of apoptosis. In particular
embodiments, the monitoring component is an agent that fluoresces at a particular
wavelength when apoptosis occurs. For example, in a preferred embodiment, caspase
activity activates green fluorescence in the monitoring component. Apoptotic cancer
cells, which have turned red as a result of being targeted by a particular signature with a
red label, turn orange while residual cancer cells remain red. Normal cells induced to
undergo apoptosis (e.g., through collateral damage), if present, will fluoresce green.
In these embodiments, fluorescent groups such as fluorescein are employed in the
monitoring component. Fluorescein is easily attached to the dendrimer surface via the
isothiocyanate derivatives, available from Molecular Probes, Inc. This allows the
nanodevices to be imaged with the cells via confocal microscopy.
Sensing of the effectiveness of the nanodevices is preferably achieved by using
fluorogenic peptide enzyme substrates. For example, apoptosis caused by the therapeutic
agents results in the production of the peptidase caspase-1 (ICE). Calbiochem sells a
number of peptide substrates for this enzyme that release a fluorescent moiety. A
particularly useful peptide for use in the present invention is:
MCA-Tyr-Glu-Val-Asp-Gly-Trp-Lys-(DNP)-NH2 (SEQ ID NO:l)
where MCA is the (7-methoxycoumarin-4-yl)acetyl and DNP is the 2,4-dinitrophenyl
group (Talanian et al., J. Biol. Chem., 272: 9677 [1997]). In this peptide, the MCA
group has greatly attenuated fluorescence, due to fluorogenic resonance energy transfer
(FRET) to the DNP group. When the enzyme cleaves the peptide between the aspartic
acid and glycine residues, the MCA and DNP are separated, and the MCA group strongly
fluoresces green (excitation maximum at 325 nm and emission maximum at 392 nm).
In preferred embodiments of the present invention, the lysine end of the peptide is
linked to the nanodevice, so that the MCA group is released into the cytosol when it is
cleaved. The lysine end of the peptide is a useful synthetic handle for conjugation
because, for example, it can react with the activated ester group of a bifunctional linker
such as Mal-PEG-OSu. Thus the appearance of green fluorescence in the target cells
produced using these methods provides a clear indication that apoptosis has begun (if the
cell already has a red color from the presence of aggregated quantum dots, the cell turns
orange from the combined colors).
Additional fluorescent dyes that find use with the present invention include, but
are not limited to, acridine orange, reported as sensitive to DNA changes in apoptotic
cells (Abrams et al., Development 117:29 [1993]) and cis-parinaric acid, sensitive to the
lipid peroxidation that accompanies apoptosis (Hockenbery et al., Cell 75:241 [1993]). It
should be noted that the peptide and the fluorescent dyes are merely exemplary. It is
contemplated that any peptide that effectively acts as a substrate for a caspase produced
as a result of apoptosis finds use with the present invention.
VI. Targeting Components
As described above, another component of the present invention is that the
nanodevice compositions are able to specifically target a particular cell type (e.g., rumor
cell). Generally, the nanodevice targets neoplastic cells through a cell surface moiety and
is taken into the cell through receptor mediated endocytosis.
Any moiety known to be located on the surface of target cells (e.g., tumor cells)
finds use with the present invention. For example, an antibody directed against such a
moiety targets the compositions of the present invention to cell surfaces containing the
moiety. Alternatively, the targeting moiety may be a ligand directed to a receptor
present on the cell surface or vice versa. Similarly, vitamins also may be used to target
the therapeutics of the present invention to a particular cell.
In some embodiments of the present invention, the targeting moiety may also
function as a signatures component. For example, tumor specific antigens including, but
not limited to, carcinoembryonic antigen, prostate specific antigen, tyrosinase, ras, a
sialyly lewis antigen, erb, MAGE-1, MAGE-3, BAGE, MN, gplOO, gp75, p97, proteinase
3, a mucin, CD81, CID9, CD63; CD53, CD38, CO-029, CA125, GD2, GM2 and
O-acetyl GD3, M-TAA, M-fetal or M-urinary find use with the present invention.
Alternatively the targeting moiety may be a tumor suppressor, a cytokine, a chemokine, a
tumor specific receptor ligand, a receptor, an inducer of apoptosis, or a differentiating
agent
Tumor suppressor proteins contemplated for targeting include, but are not limited
to, pl6, p21, p27, p53, p73, Rb, Wilms tumor (WT-1), DCC, neurofibromatosis type 1
(NF-1), von Hippel-Lindau (VHL) disease tumor suppressor, Maspin, Brush-1, BR.CA-1,
BRCA-2, the multiple tumor suppressor (MTS), gp95/p97 antigen of human melanoma,
renal cell carcinoma-associated G250 antigen, KS 1/4 pan-carcinoma antigen, ovarian
carcinoma antigen (CA125), prostate! specific antigen, melanoma antigen gp75, CD9,
CD63, CD53, CD37, R2, CD81, CO029, TI-1, L6 and SAS. Of course these are merely
exemplary tumor suppressors and it is envisioned that the present invention may be used
in conjunction with any other agent that is or becomes known to those of skill in the art
as a tumor suppressor.
In preferred embodiments of the present invention targeting is directed to factors
expressed by an oncogene. These include, but are not limited to, tyrosine kinases, both
membrane-associated and cytoplasmic forms, such as members of the Src family,
serine/threonine kinases, such as Mos, growth factor and receptors, such as platelet
derived growth factor (PDDG), SMALL GTPases (G proteins) including the ras family,
cyclin-dependent protein kinases (cdk), members of the myc family members including
c-myc, N-myc, and L-myc and bcl-2 and family members.
Cytokines, that may be targeted by the present invention include, but are not
limited to, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, ILA 1, IL-12,
IL-13, EL-14, IL-15, TNF, GMCSF, p-interferon and y-interferon. Chemokines that may
be used include, but are not limited to, MlPlα, MlPlβ, and RANTES.
Enzymes that may be targeted by the present invention include, but are not
limited to, cytosine deaminase, hypoxanthine-guanine phosphoribosyltransferase,
galactose-1-phosphate uridyltransferase, phenylalanine hydroxylase, glucocerbrosidase,
sphingomyelinase, α-L-iduronidase, glucose-6-phosphate dehydrogenase, HSV thymidine
kinase, and human thymidine kinase.
Receptors and their related ligands that find use in the context of the present
invention include, but are not limited to, the folate receptor, adrenergic receptor, growth
hormone receptor, luteinizing hormone receptor, estrogen receptor, epidermal growth
factor receptor, fibroblast growth factor receptor, and the like.
Hormones and their receptors that find use in the targeting aspect of the present
invention include, but are not limited to, growth hormone, prolactin, placental lactogen,
luteinizing hormone, foihcle-stimulating hormone, chorionic gonadotropin,
thyroid-stimulating hormone, leptin, adrenocorticotropin (ACTH), angiotensin I,
angiotensin U, β-endorphin, βmelanocyte stimulating hormone (β-MSH), cholecystokinin,
endothelin I, galanin, gastric inhibitory peptide (GIP), glucagon, insulin, amylin,
lipotropins, GLP-1 (7-37) neurophysins, and somatostatin.
In addition, the present invention contemplates that vitarnins (both fat soluble and
non-fat soluble vitamins) placed in the targeting component of the nanodevice may be
used to target cells that have receptors for, or otherwise take up these vitamins.
Particularly preferred for this aspect are the fat soluble vitamins, such as vitamin D and
its analogues, vitamin E, Vitamin A, and the like or water soluble vitamins such as
Vitamin C, and the like.
In some embodiments of the present invention, any number of cancer cell
targeting groups are attached to dendrimers. The targeting dendrimers are, in turn,
conjugated to a core dendrimer. Thus the nanodevice of the present invention is such
that it is specific for targeting cancer cells (i.e., much more likely to attach to cancer
cells and not to healthy cells). In addition, the polyvalency of dendrimers allows the
attachment of polyethylene glycol (PEG) or polyethyloxazoline (PEOX) chains to help
increase the blood circulation time and decrease the immunogenicity of the conjugates.
In preferred embodiments of the present invention, targeting groups are
conjugated to dendrimers with either short (e.g., direct coupling), medium (e.g., using
small-molecule bifunctional linkers such as SPDP, sold by Pierce Chemical Company), or
long (e.g., PEG bifunctional linkers, sold by Shearwater Polymers) linkages. Since
dendrimers have surfaces with a large number of functional groups, more than one
targeting group may be attached to each dendrimer. As a result, there are multiple
binding events between the dendrimer and the target cell. In these embodiments, the
dendrimers have a very high affinity for their target cells via this "cooperative binding"
or polyvalent interaction effect.
For steric reasons, the smaller the ligands, the more can be attached to the surface
of a dendrimer. Recently, Wiener reported that dendrimers with attached folic acid
would specifically accumulate on the surface and within tumor cells expressing the
high-affinity folate receptor (hFR) (Wiener et al., Invest. Radiol., 32:748 [1997]). The
hFR receptor is expressed or upregulated on epithelial tumors, including breast cancers.
Control cells lacking hFR showed no significant accumulation of folate-derivatized
dendrimers. Folic acid can be attached to full generation PAMAM dendrimers via a
carbodiimide coupling reaction. Folic acid is a good targeting candidate for the
dendrimers, with its small size and a simple conjugation procedure.
A larger, yet still relatively small ligand is epidermal growth factor (EGF), a
single-chain peptide with 53 amino acid residues. It has been shown that PAMAM
dendrimers conjugated to EGF with the linker SPDP bind to the cell surface of human
glioma cells and are endocytosed, accumulating in lysosomes (Casale et al., Bioconjugate
Chem., 7:7 [1996]). Since EGF receptor density is up to 100 times greater on brain
tumor cells compared to normal cells, EGF provides a useful targeting agent for these
kinds of tumors. Since the EGF receptor is also overexpressed in breast and colon
cancer, EGF may be used as a targeting agent for these cells as well. Similarly, the
fibroblast growth factor receptors (EGER) also bind the relatively small polypeptides
(FGF), and many are known to be expressed at high levels in breast rumor cell lines
(particularly FGF1, 2 and 4) (Penault-Llorca et al., Int. J. Cancer 61:170 [1995]).
In preferred embodiments of the present invention, the targeting moiety is an
antibody or antigen binding fragment of an antibody (e.g., Fab units). For example, a
well-studied antigen found on the surface of many cancers (including breast HER2
tumors) is glycoprotein pl85, which is exclusively expressed in malignant cells (Press et
al., Oncogene 5:953 [1990]). Recombinant humanized anti-HER2 monoclonal antibodies
(rhuMabHER2) have even been shown to inhibit the growth of HER2 overexpressing
breast cancer cells, and are being evaluated (in conjunction with conventional
chemotherapeutics) in phase III clinical trials for the treatment of advanced breast cancer
(Pegrarn et al., Proc. Am. Soc. Clin. Oncol., 14:106 [1995]). Park and Papahadjopoulos
have attached Fab fragments of rhuMabHER2 to small unilamellar liposomes, which then
can be loaded with the chemotherapeutic doxorubicin (dox) and targeted to HER2
overexpressing tumor xenografts (Park et al., Cancer Lett., 118:153 [1997] and Kirpotin
et al., Biochem., 36:66 [1997]). These dox-loaded "immunoliposomes" showed increased
cytotoxicity against tumors compared to corresponding non-targeted dox-loaded
liposomes or free dox, and decreased systemic toxicity compared to free dox.
Antibodies can be generated to allow for the targeting of antigens or immunogens
(e.g., tumor, tissue or pathogen specific antigens) on various biological targets (e.g.,
pathogens, tumor cells, normal tissue). Such antibodies include, but are not limited to
polyclonal., monoclonal., chimeric, single chain, Fab fragments, and an Fab expression
library.
In some preferred embodiments, the antibodies recognize tumor specific epitopes
(e.g., TAG-72 (Kjeldsen et al., Cancer Res. 48:2214-2220 [1988]; U.S. Pat Nos.
5,892,020; 5,892,019; and 5,512,443); human carcinoma antigen (U.S. Pat Nos.
5,693,763; 5,545,530; and 5,808,005); TP1 and TP3 antigens from osteocarcinoma cells
(U.S. Pat No. 5,855,866); Thomsen-Friedenreich (TF) antigen from adenocarcinoma
cells (U.S. Pat. No. 5,110,911); "KC-4 antigen" from human prostrate adenocarcinoma
(U.S. Pat. Nos. 4,708,930 and 4,743,543); a human colorectal cancer antigen (U.S. Pat.
No. 4,921,789); CA125 antigen from cystadenocarcinoma (U.S. Pat. No. 4,921,790); DF3
antigen from human breast carcinoma (U.S. Pat. Nos. 4,963,484 and 5,053,489); a human
breast tumor antigen (U.S. Pat. No. 4,939,240); p97 antigen of human melanoma (U.S.
Pat. No. 4,918,164); carcinoma or orosomucoid-related antigen (CORA)(U.S. Pat. No.
4,914,021); a human pulmonary carcinoma antigen that reacts with human squamous cell
lung carcinoma but not with human small cell lung carcinoma (U.S. Pat. No. 4,892,935);
T and Tn haptens in glycoproteins of human breast carcinoma (Springer et al.,
Carbohydr. Res. 178:271-292 [1988]), MSA breast carcinoma glycoprotein termed
(Tjandra et al., Br. J. Surg. 75:811-817 [1988]); MFGM breast carcinoma antigen (Ishida
et al., Tumor Biol. 10:12-22 [1989]); DU-PAN-2 pancreatic carcinoma antigen (Lan et
al., Cancer Res. 45:305-310 [1985]); CA125 ovarian carcinoma antigen (Hanisch et al.,
Carbohydr. Res. 178:29-47 [1988]); YH206 lung carcinoma amigen (Hinoda et al.,
(1988) Cancer J. 42:653-658 [1988]). Each of the foregoing references are specifically
incorporated herein by reference.
In other preferred embodiments, the antibodies recognize specific pathogens (e.g.,
Legionella peomophilia, Mycobacterium tuberculosis, Clostridium tetani, Hemophilus
influenzae, Neisseria gonorrhoeae, Treponema pallidum, Bacillus anthracis, Vibrio
cholerae, Borrelia burgdorferi, Cornebacterium diphtheria, Staphylococcus aureus,
human papilloma virus, human immunodeficiency virus, rubella virus, polio virus, and
the like).
Various procedures known in the art are used for the production of polyclonal
antibodies. For the production of antibody, various host animals can be immunized by
injection with the peptide corresponding to the desired epitope including but not limited
to rabbits, mice, rats, sheep, goats, etc. In a preferred embodiment, the peptide is
conjugated to an immunogenic carrier (e.g., diphtheria toxoid, bovine serum albumin
(BSA), or keyhole limpet hemocyanin (KLH)). Various adjuvants are used to increase
the immunological response, depending on the host species, including but not limited to
Freund's (complete and incomplete), mineral gels such as aluminum hydroxide, surface
active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil
emulsions, keyhole limpet hemocyanins, dinitrophenol, and potentially useful human
adjuvants such as BCG (Bacille Calmette-Guerin) and Corynebacterium parvum.
For preparation of monoclonal antibodies, any technique1 that provides for the
production of antibody molecules by continuous cell lines in culture may be used (See
e.g., Harlow and Lane, Antibodies: A Laboratory Manual., Cold Spring Harbor
Laboratory Press, Cold Spring Harbor, NY). These include, but are not limited to, the
hybridoma technique originally developed by Kohler and Milstdin (Kohler and Milstein,
Nature 256:495-497 [1975]), as well as the trioma technique, the human B-cell
hybridoma technique (See e.g., Kozbor et al. Immunol. Today 4:72 [1983]), and the
EBV-hybridoma technique to produce human monoclonal antibodies (Cole et al., in
Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96 [1985]).
In an additional embodiment of the invention, monoclonal antibodies can be
produced in germ-free animals utilizing recent technology {See e.g., PCT/US90/02545).
According to the invention, human antibodies may be used and can be obtained by using
human hybridomas (Cote et al., Proc. Natl. Acad. Sci. U.S.A.80:2026-2030 [1983]) or by
transforming human B cells with EBV virus in vitro (Cole et al., in Monoclonal
Antibodies and Cancer Therapy, Alan R. Liss, pp. 77-96 [1985]).
According to the invention, techniques described for the production of single
chain antibodies (U.S. Patent 4,946,778; herein incorporated by reference) can be adapted
to produce specific single chain antibodies. An additional embodiment of the invention
utilizes the techniques described for the construction of Fab expression libraries (Huse et
al., Science 246:1275-1281 [1989]) to allow rapid and easy identification of monoclonal
Fab fragments with the desired specificity.
Antibody fragments that contain the idiotype (antigen binding region) of the
antibody molecule can be generated by known techniques. For example, such fragments
include but are not limited to: the F(ab')2 fragment that can be produced by pepsin
digestion of the antibody molecule; the Fab' fragments that can be generated by reducing
the disulfide bridges of the F(ab')2 fragment, and the Fab fragments that can be
generated by treating the antibody molecule with papain and a reducing agent.
In the production of antibodies, screening for the desired antibody can be
accomplished by techniques known in the art (e.g., radioimmunoassay, ELISA
(enzyme-linked immunosorbant assay), "sandwich" immunoassays, immunoradiometric
assays, gel diffusion precipitin reactions, immunodiffusion assays, in situ immunoassays
(using colloidal gold, enzyme or radioisotope labels, for example), Western Blots,
precipitation reactions, agglutination assays {e.g., gel agglutination assays,
hemagglutination assays, etc.), complement fixation assays, immunofluorescence assays,
protein A assays, and immunoelectrophoresis assays, etc.).
The dendrimer systems of the present invention have many advantages over
liposomes, such as their greater stability, better control of their size and polydispersity,
and generally lower toxicity and immunogenicity (See e.g., Duncan et al., Polymer
Preprints 39:180 [1998]). Thus, in some embodiments of the present invention,
anti-HER2 antibody fragments, as well as other targeting antibodies are conjugated to
dendrimers, as targeting agents for the nanodevices of the present inventioa
For breast cancer, the cell surface may be targeted with folic acid, EGF, FGF, and
antibodies (or antibody fragments) to the tumor-associated antigens MUC1, cMet receptor
and CD56 (NCAM). Once internalized into the cell, the nanodevice binds (via
conjugated antibodies) to HER2, MUC1 or mutated p53.
The bifunctional linkers SPDP and SMCC and the longer Mal-PEG-OSu linkers
are particularly useful for antibody-dendrimer conjugation. In addition, many tumor cells
contain surface lectins that bind to oligosaccharides, with specific recognition arising
chiefly from the terminal carbohydrate residues of the latter (Sharon and Lis, Science
246:227 [1989]). Attaching appropriate monosaccharides to nonglycosylated proteins
such as BSA provides a conjugate that binds to tumor lectin much more tightly than the
free monosaccharide (Monsigny et al., Biochemie 70:1633 [1988]).
Mannosylated PAMAM dendrimers bind mannoside-binding lectin up to 400 more
avidly than monomelic mannosides (Page and Roy, Bioconjugate Chem., 8:714 [1997]).
Sialylated dendrimers and other dendritic polymers bind to and inhibit a variety of
sialate-binding viruses both in vitro and in vivo. By conjugating multiple
monosaccharide residues {e.g., α-galactoside, for galactose-binding cells) to dendrimers,
polyvalent conjugates are created with a high affinity for the corresponding type of tumor
cell. The attachment reaction are easily carried out via reaction of the terminal amines
with commercially-available α-galactosidyl-phenylisothiocyanate. The small size of the
carbohydrates allows a high concentration to be present on the dendrirner surface.
A very flexible method to identify and select appropriate peptide targeting groups
is the phage display technique (See e.g., Cortese et al., Curr. Opin. Biotechol., 6:73
[1995]), which can be conveniently carried out using commercially available kits. The
phage display procedure produces a large and diverse combinatorial library of peptides
attached to the surface of phage, which are screened against immobilized surface
receptors for tight binding. After the tight-binding, viral constructs are isolated and
sequenced to identify the peptide sequences. The cycle is repeated using the best
peptides as starting points for the next peptide library. Eventually, suitably high-affinity
peptides are identified and then screened for biocompatibility and target specificity. In
this way, it is possible to produce peptides that can be conjugated to dendrimers,
producing multivalent conjugates with high specificity and affinity for the target cell
receptors (e.g., tumor cell receptors) or other desired targets.
.Related to the targeting approaches described above is the "pretargeting" approach
(See e.g., Goodwin and Meares, Cancer (suppl.) 80:2675 [1997]). An example of this
strategy involves initial treatment of the patient with conjugates of tumor-specific
monoclonal antibodies and streptavidin. Remaining soluble conjugate is removed from
the bloodstream with an appropriate biotinylated clearing agent. When the
tumor-localized conjugate is all that remains, a radiolabeled, biotinylated agent is
introduced, which in turn localizes at the tumor sites by the strong and specific
biotin-streptavidin interaction. Thus, the radioactive dose is maximized in dose
proximity to the cancer cells and minimized in the rest of the body where it can harm
healthy cells.
It has been shown that if streptavidin molecules bound to a polystyrene well are
first treated with a biotinylated dendrimer, and then radiolabeled streptavidinis
introduced, up to four of the labeled streptavidin molecules are bound per
polystyrene-bound streptavidin (Wilbur et al., Bioconjugate Chem., 9:813 [1998]). Thus,
biotinylated dendrimers may be used in the methods of the present invention, acting as a
polyvalent receptor for the radiolabel in vivo, with a resulting amplification of the
radioactive dosage per bound antibody conjugate. In the preferred embodiments of the
present invention, one or more multiply-biotinylated module(s) on the clustered
dendrimer presents a polyvalent target for radiolabeled or boronated (Barth et al., Cancer
Investigation 14:534 [1996]) avidin or streptavidin, again resulting in an amplified dose .
of radiation for the tumor cells.
Dendrimers and clustered dendrimers may also be used as clearing agents by, for
example, partially biotinylating a dendrimer that has a polyvalent galactose or mannose
surface. The conjugate-clearing agent complex would then have a very strong affinity
for the corresponding hepatocyte receptors.
In other embodiments of the present invention, an enhanced permeability and
retention (EPR) method is used in targeting. The enhanced permeability and retention
(EPR) effect is a more "passive" way of targeting tumors (See, Duncan and Sat, Ann.
Oncol., 9:39 [1998]). The EPR effect is the selective concentration of macromolecules
and small particles in the tumor microenvironment, caused by the hyperpermeable
vasculature and poor lymphatic drainage of tumors. The dendrimer compositions of the
present invention provide ideal polymers for this application, in that they are relatively
rigid, of narrow polydispersity, of controlled size and surface chemistry, and have
interior "cargo" space that can carry and then release antitumor drugs. In fact, PAMAM
dendrimer-platinates have been shown to accumulate in solid rumors (Pt levels about 50
times higher than those obtained with cisplatin) and have in vivo activity in solid tumor
models for which cisplatin has no effect (Malik et al., Proc. Int'l. Symp. Control. Rel.
Bioact. Mater., 24:107 [1997] and Duncan et al., Polymer Preprints 39:180 [1998]).
The targeting moieties of the present invention may recognize a variety of other
epitopes on biological targets (e.g., on pathogens). In some embodiments, molecular
recognition elements are incorporated to recognize, target or detect a variety of
pathogenic organisms including, but not limited to, sialic acid to target HIV (Wies et al.,
Nature 333: 426 [1988]), influenza (White et al., Cell 56: 725 [1989]), Chlamydia
(Infect. Imm. 57: 2378 [1989]), Neisseria meningitidis, Streptococcus suis, Salmonella,
mumps, newcastle, and various viruses, including reovirus, Sendai virus, and myxovirus;
and 9-OAC sialic acid to target coronavirus, encephalomyelitis virus, and rotavirus; non-
sialic acid glycoproteins to detect cytomegalovirus (Virology 176: 337 [1990]) and
measles virus (Virology 172: 386 [1989]); CD4 (Khatzman et al., Nature 312: 763
[1985]), vasoactive intestinal peptide (Sacerdote et al., J. of Neuroscience Research 18:
102 [1987]), and peptide T (Ruff et al., FEBS Letters 211: 17 [1987]) to target HTV;
epidermal growth factor to target vaccinia (Epstein et al., Nature 318: 663 [1985]);
acetylcholine receptor to target rabies (Lentz et al., Science 215: 182 [1982]); Cd3
complement receptor to target Epstein-Barr virus (Carel et al., J. Biol. Chem. 265: 12293
[1990]); p-adrenergic receptor to target reovirus (Co et al., Proc. Natl. Acad. Sci. 82:
1494 [1985]); ICAM-1 (Marlin et al., Nature 344: 70 [1990]), N-CAM, and myelin-
associated glycoprotein MAb (Shephey et al., Proc. Natl. Acad, Sci. 85: 7743 [1988]) to
target rninovirus; polio virus receptor to target polio virus (Mendelsohn et al., Cell 56:
855 [1989]); fibroblast growth factor receptor to target herpes virus (Kaner et al.,
Science 248: 1410 [1990]); oligomannose to target Escherichia coli; ganglioside GMI to
target Neisseria meningitidis', and antibodies to detect a broad variety of pathogens (e.g.,
Neisseria gonorrhoeae, V. vulnificus, V. parahaemolyticus, V. cholerae, and V.
alginolyticus).
In some embodiments of the present invention, the targeting moities are
preferably nucleic acids (e.g., RNA or DNA). In some embodiments, the nucleic acid
targeting moities are designed to hybridize by base pairing to a particular nucleic acid
(e.g., chromosomal DNA, mRNA, or ribosomal RNA). In other embodiments, the
nucleic acids bind a ligand or biological target. Nucleic acids that bind the following
proteins have been identified: reverse transcriptase, Rev and Tat proteins of HTV (Tuerk
et al., Gene 137(l):33-9 [1993]); human nerve growth factor (Binkley et al., Nuc. Acids
Res. 23(16):3198-205 [1995]); and vascular endothelial growth factor (Jellinek et al.,
Biochem. 83(34): 10450-6 [1994]). Nucleic acids that bind ligands are preferably
identified by the SELEX procedure (See e.g., U.S. Pat. Nos. 5,475,096; 5,270,163; and
5,475,096; and in PCT publications WO 97/38134, WO 98/33941, and WO 99/07724, all
of which are herein incorporated by reference), although many methods are known in the
art.
VII. Synthesis and Conjugation
The present section provides a description of the synthesis and formation of the
individual components (i.e., individual dendrimers containing one or more of the
components described above) of the nanodevice and the conjugation of such components
into a nanodevice (e.g., the conjugation of one or more such dendrimers to a core
dendrimer).
In preferred embodiments of the present invention, the preparation of PAMAM
dendrimers is performed according to a typical divergent (building up the macromolecule
from an initiator core) synthesis. It involves a two-step growth sequence that consists of
a Michael addition of amino groups to the double bond of methyl acrylate (MA)
followed by the amidation of the resulting terminal carbomethoxy, -(CO2CH3) group,
with ethylenediamine (EDA). When ammonia is used as the initiator core reagent, this
synthesis may be represented by reactions shown in Figure 7.
In the first step of this process, ammonia is allowed to react under an inert
nitrogen atmosphere with MA (molar ratio: 1:4.25) at 47 °C for 48 hours. The resulting
compound is referred to as generation = 0, the star-branched PAMAM tri-ester. The next
step involves reacting the tri-ester with an excess of EDA to produce the star-branched
PAMAM tri-amine (G=0). This reaction is performed under an inert atmosphere
(nitrogen) in methanol and requires 48 hours at 0 °C for completion. Reiteration of this
Michael addition and amidation sequence produces generation = 1.
Preparation of this tri-amine completes the first full cycle of the divergent
synthesis of PAMAM dendrimers. Repetition of this reaction sequence results in the
synthesis of larger generation (G=l-5) dendrimers (i.e., ester- and amine-terminated
molecules, respectively). For example, the second iteration of this sequence produces
generation 1, with an hexa-ester and hexa-amine surface, respectively. The same
reactions are performed in the same way as for all subsequent generations from 1 to 9,
building up layers of branch cells giving a core-shell architecture with precise molecular
weights and numbers of terminal groups as shown above. Carboxylate-surfaced
dendrimers can be produced by hydrolysis of ester-terminated PAMAM dendrimers, or
reaction of succinic anhydride with amine-surfaced dendrimers (e.g., full generation
PAMAM, POP AM or POPAM-PAMAM hybrid dendrimers).
Various dendrimers can be synthesized based on the core structure that initiates
the polymerization process. These core structures dictate several important characteristics
of the dendrimer molecule such as the overall shape, density, and surface functionality
(Tomalia et al., Angew. Chem. Int. Ed. Engl., 29:5305 [1990]). Spherical dendrimers
derived from ammonia possess trivalent initiator cores, whereas EDA is a tetra-valent
initiator core. Recently, rod-shaped dendrimers have been reported which are based upon
linear poly(ethyleneimine) cores of varying lengths the longer the core, the longer the rod
(Yin et al., J. Am. Chem. Soc, 120:2678 [1998]).
The dendrimers may be characterized for size and uniformity by any suitable
analytical techniques. These include, but are not limited to, atomic force microscopy
(AFM), electrospray-ionization mass spectroscopy, MALDI-TOF mass spectroscopy, 13C
nuclear magnetic resonance spectroscopy, high performance liquid chromatography
(HPLC) size exclusion chromatography (SEC) (equipped with multi-angle laser light
scattering, dual UV and refractive index detectors), capillary electrophoresis and get
electrophoresis. These analytical methods assure the uniformity of the dendrimer
population and are important in the quality control of dendrimer production for eventual
use in in vivo applications. Most importantly, extensive work has been performed with
dendrimers showing no evidence of toxicity when administered intravenously (Roberts et
al., J. Biomed. Mater. Res., 30:53 [1996] and Bourne et al., J. Magnetic Resonance
Imaging, 6:305 [1996]).
To produce a single dendritic device possessing the various functional modules
required for active sensing, targeting, imaging and therapeutic delivery, multiple
PAMAM dendrimer modules, each with an individual differentiated function are
covalently bound to form a single device. This involves the synthesis of separate
conjugates or nanocomposites for each of the required activities (e.g., one dendrimer
conjugate for sensing, one for targeting and another for therapeutic carrier). These
different dendrimers are then self-assembled and covalently linked in a manner that
yields a single therapeutic device. In certain embodiments, one dendrimer acts as a core
around which other dendrimers are covalently (i.e., "clustered dendrimers"). In preferred
embodiments, the core dendrimer is a POPAM dendrimer, while the outer dendrimers are
PAMAM dendrimers. In yet other embodiments, dendrimers may be complexed to one
another without a core dendrimer (e.g., four dendrimers covalently linked to one another
in a linear chain).
In one preferred embodiments of the present invention the formation of clustered
dendrimers involves the formation of amide bonds between the core and exterior
dendrimers using the ester aminolysis technique. The ester aminolysis technique involves
reacting various poly(arnidoamine) PAMAM dendrimer core reagents with an excess of
ester terminated PAMAM dendrimer shell reagents in methanol at 40 °C (See e.g.,
Uppuluri et al., PMSE 80:55 [1999]). In an alternative embodiment, water is employed
as the reaction medium. This method involves the self-assembly of amine terminated
core reagents with an excess of carboxylate shell reagent followed by addition of a
coupling agent (i.e., carboimide) to produce arninde linkages between the core and the
shell components. These reactions take place at room temperature. Such embodiments
are preferred when the reactions are conducted in the presence of biomolecules such as
antibodies.
The first step in the aqueous synthesis of these molecules involves self-assembly
of the shell dendrimer molecules around a core dendrimer molecule, resulting in the
efficient (i.e., maximum) packing of shell molecules around the core. The self-assembled
cluster, as shown in Figure 5, is representative of the precursor used to make the
covalently bonded core shell clustered dendrimer. In the next step, using a coupling
reagent (such as EDC, a carbodiimide reagent), the core and shell molecules are
covalently linked as shown in Figure 6. The reaction progress is monitored by size
exclusion chromatography (SEC) and the loss of carboxylate functionality in the infrared
region (FTIR) as well as by 1H/13C NMR and gel electrophoresis. The reaction is
normally complete within an hour when run at room temperature.
In some embodiments of the present invention, the size and shape of these higher
molecular weight products is measured and compared to individual dendrimers by atomic
force microscopy (AFM) and size exclusion chromatography SEC. These techniques
demonstrate that core-shell dendrimers are indeed formed. Additional evidence is
obtained, as desired, by gel electrophoresis, in which a higher molecular weight product
is evident when the reaction is complete. The absolute molecular weight of the clustered
dendrimer is determined by MALDI-TOF mass spectroscopy or by SEC equipped with a
multi-angle laser light scattering detector (MALLS).
The clustered dendrimer molecules formed by this method have narrow
polydispersity by SEC (similar to that of large dendrimers). It takes about 3-4 weeks to
convert PAMAM dendrimers of generation 6 to generation 9, but only about 1 day to
synthesize clustered dendrimers with similar size, acceptable dispersity and shape
(including purification procedures).
For the multi-function clustered dendrimer, any cross-linking reaction problems
with the functional groups on the exterior dendrimer modules are circumvented by using
standard protecting groups on the side chains that are reacting. Another solution is to use
bifunctional linker strategies, e.g., first, reacting the surface of the core amino-surfaced
dendrimer with 2-iminothiolane to generate a thiol surface, then reacting the product with
maleimide linker groups on the shell dendrimers.
As discussed above, in some embodiments of the present invention a core-shell
structure is used to assemble dendritic polymer components into a single molecular
complex (See e.g., Figure 10). This allows one to place each component of the
nanodevice on a different polymer and assemble them as a single, supramolecular
assembly. The unique aspect of this technology is that the core-shell configuration
directs and limits the assembly; the larger the core and the smaller the shell molecules,
the greater the number of shell dendrimers can associate with a core. For example, the
core of a cluster can be generation 7, amine surfaced PAMAM dendrimer; with an
approximate molecular weight of 110kDa, a 7 nm diameter and 512 surface primary
amines (Tomalia et at, Angew. Chem. Int. Ed., 29:138 [1990]). The shell might be made
up of generation 5 carboxyl-surfaced PAMAM dendrimer, with an approximate molecular
weight of 27kDa, a 5 nm diameter and 128 surface carboxyl groups. This would lead to
the self-assembly (if performed in an excess of E5) of a supra-molecular complex where
an average of 12 E5 molecules surround an E7 core. Figure 10 shows that steric
hindrance limits the number of associated shell polymers that bind to the core.
There may be problems in attempting to assemble dendrimer-based nanodevices
using this approach with multifunctional groups using covalently linked PAMAM
dendrimers. For example, in some embodiments, the self-assembly of these components
requires different charges on the core and surface polymers. This leaves excess charge
on the shell polymer that may inhibit the function of the device, particularly as relates to
targeting and internalizing into cells. Also important is that after the polymers
self-assembly, in some embodiments, they are covalently linked. The chemistry involved
in this step can affect the functional subunits of the nanodevice and in some cases
destroy them. Thus, an alternative technique should be employed to assemble the
supramolecular complex.
In some embodiments of the present invention, individual dendrimer components
of the multi-function clustered dendrimer are assembled though the use of linkers. For
example, in some embodiments, shell dendrimers are attached to a core dendrimer
through linker groups using covalent or non-covalent interactions. One illustrative
example of such linking is demonstrated by the use of nucleic acid linkers, which provide
a number of advantages.
Oligonucleotides are a powerful tool to assemble molecules in desired structural
arrangements due to the ease with which they can be conjugated to other materials, the
ability to hybridize with another oligonucleotide of complementary base specificity, the
programmability of the sequence, and the stiffness of the resulting duplex structure.
Using this concept, supramolecular core-shell structures using dendrimers and
complementary oligonucleotides were created during the development of the present
invention. These structures have a number of advantages over those produced with the
prior assembly techniques. For example, they are self assembling at low temperature
without harsh chemicals, they provide the ability to specifically couple different subunits
based on different oligonucleotide complementarities, and they provide the ability to
design nuclease digestion sites or other cleavable sites into the oligonucleotide couplers
to make the complex "biodegradable" and allow the subunits to be excreted through the
kidney (e.g., they may be designed to fall apart once they've reached the target site, such
that the smaller fragments may be excreted through the kidney). Figure 11 shows a
schematic dendrimer complex assembled using nucleic acid linkers. A method for
preparing the complexes is provided in Example 5.
The present invention is not limited by the nature of the nucleic acid used as the
linker group. In some preferred embodiments, the nucleic acid attached to a dendrimer
does not contain intrastrand secondary structure. However, in some embodiments
secondary structure may be used to provide a desired function or property (e.g., stability,
cleavage recognition site, etc.). The length of the nucleic acid linker may be selected to
provide a desired distance between the core dendrimer and the shell dendrimers. In
some embodiments of the present invention, nucleic acids are modified to enhance
stability (e.g., in the bloodstream) and/or to facilitate entry into cells. Methods are
known in the art for making such modifications. In some embodiments, the nucleic acid
molecules are labeled to allow detection or localization of the assemblies.
Once nucleic acid linkers are attached to the dendrimers, the dendrimer complexes
may be assembled and analyzed (e.g., to assure the structures have appropriate
conformations). Because of the small size of these materials, a preferred method of
characterizing the assembled complexes is atomic force microscopy. Use of atomic force
microscopy clearly demonstrated the presence of dendrimer supramolecular assembles
that are regular combinations of three and four modules (i.e., comprised a core and
multiple shell dendrimers). Analysis of a single cluster demonstrated that the distance
between the two components was 21 nm; almost exactly the theoretical distance predicted
from the length of the oligonucleotide hybrid. This analysis was then applied to a
population of the molecules, and all were found to have distances of approximately 20
nm between the components. This documented the uniformity of the supramolecular
structures that were developed. Given the fact that these supramolecular assemblies can
connect components in a consistent manner, they can be used for almost any type of
combined delivery of material from vaccine components to drugs to imaging agents.
VIE Evaluation of Anti-Tumor Efficacy and Toxicity of Nanodevice
The anti-tumor effects of various therapeutic agents on cancer cell lines and
primary cell cultures may be evaluated using the nanodevices of the present invention.
For example, in preferred embodiments, assays are conducted, in vitro, using established
tumor cell line models or primary culture cells. The use of fresh tumor cells (as opposed
to cultured lines) is preferable for confirmation of toxicity testing and efficacy because it
allows more relevant determinations without artifacts induced by tissue culture (e.g.,
tumor cell selection, etc.).
A. Quantifying the Induction of Apoptosis of Human Tumor
Cells In vitro
In an exemplary embodiment of the present invention, the nanodevices of the
present invention are used to assay apoptosis of human tumor cells in vitro. Testing for
apoptosis in the cells determines the efficacy of the therapeutic agent. Multiple aspects
of apoptosis can and should be measured. These aspects include those described above,
as well as aspects including, but are not limited to, measurement of phosphatidylserine
(PS) translocation from the inner to outer surface of plasma membrane, measurement of
DNA fragmentation, detection of apoptosis related proteins, and measurement of
Caspase-3 activity.
B. In Vitro Toxicology
In some embodiments of the present invention, to gain a general perspective into
the safety of a particular nanodevice platform or component of that system, toxicity
testing is performed. Toxicological information may be derived from numerous sources
including, but not limited to, historical databases, in vitro testing, and in vivo animal
studies.
In vitro toxicological methods have gained popularity in recent years due to
increasing desires for alternatives to animal experimentation and an increased perception
to the potential ethical., commercial., and scientific value. In vitro toxicity testing systems
have numerous advantages including improved efficiency, reduced cost, and reduced
variability between experiments. These systems also reduce animal usage, eliminate
confounding systemic effects (e.g., immunity), and control environmental conditions.
Although any in vitro testing system may be used with the present invention, the
most common approach utilized for in vitro examination is the use of cultured cell
models. These systems include freshly isolated cells, primary cells, or transformed cell
cultures. Cell culture as the primary means of studying in vitro toxicology is
advantageous due to rapid screening of multiple cultures, usefulness in identifying and
assessing toxic effects at the cellular, subcellular, or molecular level. In vitro cell culture
methods commonly indicate basic cellular toxicity through measurement of membrane
integrity, metabolic activities, and subcellular perturbations. Commonly used indicators
for membrane integrity include cell viability (cell count), clonal expansion tests, trypan
blue exclusion, intracellular enzyme release {e.g., lactate dehydrogenase), membrane
permeability of small ions (K1, Ca24), and intracellular accumulation of small molecules
(e.g., 51Cr, succinate). Subcellular perturbations include monitoring mitochondrial
enzyme activity levels via, for example, the MTT test, determining cellular adenine
triphosphate (ATP) levels, neutral red uptake into lysosomes, and quantification of total
protein synthesis. Metabolic activity indicators include glutathione content, lipid
peroxiidation, and lactate/pyruvate ratio.
C. MTT assay
The MTT assay is a fast, accurate, and reliable methodology for obtaining cell
viability measurements. The MTT assay was first developed by Mosmann (Mosmann, J.
Immunol. Meth., 65:55 [1983]). It is a simple colorimetric assay numerous laboratories
have utilized for obtaining toxicity results {See e.g., Kuhlmann et al., Arch. Toxicol.,
72:536 [1998]). Briefly, the mitochondria produce ATP to provide sufficient energy for
the cell. In order to do this, the mitochondria metabolize pyruvate to produce acetyl
CoA. Within the mitochondria, acetyl CoA reacts with various enzymes in the
tricarboxylic acid cycle resulting in subsequent production of ATP. One of the enzymes
particularly useful in the MTT assay is succinate dehydrogenase. MTT
(3-(4,5-dimethylthiazol-2-yi)-2 diphenyl tetrazolium bromide) is a yellow substrate that is
cleaved by succinate dehydrogenase forming a purple formazan product The alteration
in pigment identifies changes in mitochondria function.. Nonviable cells are unable to
produce formazan, and therefore, the amount produced directly correlates to the quantity
of viable cells. Absorbance at 540 nm is utilized to measure the amount of formazan
product.
The results of the in vitro tests can be compared to in vivo toxicity tests in order
to extrapolate to live animal conditions. Typically, acute toxicity from a single dose of
the substance is assessed. Animals are monitored over 14 days for any signs of toxicity
(increased temperature, breathing difficulty, death, etc). Traditionally, the standard of
acute toxicity is the median lethal dose (LD50), which is the predicted dose at which half
of the treated population would be killed. The determination of this dose occurs by
exposing test animals to a geometric series of doses under controlled conditions. Other
tests include subacute toxicity testing, which measures the animal's response to repeated
doses of the nanodevice for no longer than 14 days. Sub-chronic toxicity testing involves
testing of a repeated dose for 90 days. Chronic toxicity testing is similar to subchronic
testing but may last for over a 90-day period. In vivo testing can also be conducted to
determine toxicity with respect to certain tissues. For example, in some embodiments of
the present invention tumor toxicity (i.e., effect of the compositions of the present
invention on the survival of tumor tissue) is determined (e.g., by detecting changes in the
size and/or growth of tumor tissues).
IX. Gene Therapy Vectors
In particular embodiments of the present invention, the nanodevice compositions
comprise transgenes for delivery and expression to a target cell or tissue, in vitro, ex
vivo, or in vivo. In such embodiments, rather than containing the actual protein, the
dendrimer complex comprises an expression vector construct containing, for example, a
heterologous DNA encoding a gene of interest and the various regulatory elements that
facilitate the production of the particular protein of interest in the target cells.
In some embodiments, the gene is a therapeutic gene that is used, for example, to
treat cancer, to replace a defective gene, or a marker or reporter gene that is used for
selection or monitoring purposes. In the context of a gene therapy vector, the gene may
be a heterologous piece of DNA. The heterologous DNA may be derived from more
than one source (i.e., a multigene construct or a fusion protein). Further, the
heterologous DNA may include a regulatory sequence derived from one source and the
gene derived from a different source.
Tissue-specific promoters may be used to effect transcription in specific tissues or
cells so as to reduce potential toxicity or undesirable effects to non-targeted tissues. For
example, promoters such as the PSA, probasin, prostatic acid phosphatase or
prostate-specific glandular kallikrein (hK2) may be used to target gene expression in the
prostate. Similarly, promoters may be used to target gene expression in other tissues
(e.g., insulin, elastin amylase, pdr-1, pdx-1 and glucokinase promoters target to the
pancreas; albumin PEPCK, HBV enhancer, alpha fetoproteinapolipoprotein C, alpha-1
antitrypsin, vitellogenin, NF-AB and transthyretin promoters target to the liver; myosin H
chain, muscle creatine kinase, dystrophin, calpain p94, skeletal alpha-actin, fast troponin
1 promoters target to skeletal muscle; keratin promoters target the skin; sm22 alpha;
SM-α-actin promoters target smooth muscle; CFTR; human cytokeratin 18 (K18);
pulmonary surfactant proteins A, B and Q CC-10; P1 promoters target lung tissue;
endothelin-1; E-selectin; von Willebrand factor; KDR/flk-1 target the endothelium;
tyrosinase targets melanocytes).
The nucleic acid may be either cDNA or genomic DNA. The nucleic acid can
encode any suitable therapeutic protein. Preferably, the nucleic acid encodes a tumor
suppressor, cytokine, receptor, inducer of apoptosis, or differentiating agent. The nucleic
acid may be an antisense nucleic acid. In such embodiments, the antisense nucleic acid
may be incorporated into the nanodevice of the present invention outside of the context
of an expression vector.
In preferred embodiments, the nucleic acid encodes a tumor suppressor, cytokines,
receptors, or inducers of apoptosis. Suitable tumor suppressors include BRCA1, BRCA2,
C-CAM, pl6, p211 p53, p73, or Rb. Suitable cytokines include GMCSF, IL-1, IL-2,
IL-3, IL-4, IL-5, IL6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15,
βMnteferon, γ-interferon, or TNF. Suitable receptors include CFTR, EGFR, estrogen
receptor, IL-2 receptor, or VEGFR. Suitable inducers of apoptosis include AdE1B, Bad,
Bak, Bax, Bid, Bik, Bim, Harakiri, or ICE-CED3 protease.
X. Methods of Combined Therapy
Tumor cell resistance to DNA damaging agents represents a major problem in
clinical oncology. The nanodevices of the present invention provide means of
ameliorating this problem by effectively administering a combined therapy approach.
However, it should be noted that traditional combination therapy may be employed in
combination with the nanodevices of the present invention. For example, in some
embodiments of the present invention, nanodevices may be used before, after, or in
combination with the traditional therapies.
To kill cells, inhibit cell growth, or metastasis, or angiogenesis, or otherwise
reverse or reduce the malignant phenotype of tumor cells using the methods and
compositions of the present invention in combination therapy, one contacts a "target" cell
with the nanodevices compositions described herein and at least one other agent. These
compositions are provided in a combined amount effective to kill or inhibit proliferation
of the cell. This process may involve contacting the cells with the immunotherapeutic
agent and the agent(s) or factor(s) at the same time. This may be achieved by contacting
the cell with a single composition or pharmacological formulation that includes both
agents, or by contacting the cell with two distinct compositions or formulations, at the
same time, wherein one composition includes, for example, an expression construct and
the other includes a therapeutic agent
Alternatively, the nanodevice treatment may precede or follow the other agent
treatment by intervals ranging from minutes to weeks. In embodiments where the other
agent and immunotherapy are applied separately to the cell, one would generally ensure
that a significant period of time did not expire between the time of each delivery, such
that the agent and nanodevice would still be able to exert an advantageously combined
effect on the cell. In such instances, it is contemplated that cells are contacted with both
modalities within about 12-24 hours of each other and, more preferably, within about
6-12 hours of each other, with a delay time of only about 12 hours being most preferred.
In some situations, it may be desirable to extend the time period for treatment
significantly, however, where several days (2 to 7) to several weeks (1 to 8) lapse
between the respective administrations.
In some embodiments, more than one administration of the immunotherapeutic
composition of the present invention or the other agent are utilized. Various
combinations may be employed, where nanodevice is "A" and the other agent is "B", as
exemplified below:
A/B/A, B/A/B, B/B/A, A/A/B, B/A/A, A/B/B, B/B/B/A, B/B/A/B,
A/A/B/B, A/B/A/B, A/B/B/A, B/B/A/A, B/A/B/A, B/A/A/B, B/B/B/A,
A/A/A/B, B/A/A/A, A/B/A/A, A/A/B/A, A/B/B/B, B/A/B/B, B/B/A/B
Other combinations are contemplated. Again, to achieve cell killing, both agents are
delivered to a cell in a combined amount effective to kill or disable the cell.
Other factors that may be used in combination therapy with the nanodevices of
the present invention include, but are not limited to, factors that cause DNA damage such
as y-rays, X-rays, and/or the directed delivery of radioisotopes to tumor cells. Other
forms of DNA damaging factors are also contemplated such as microwaves and
UV-irradiation. Dosage ranges for X-rays range from daily doses of 50 to 200 roentgens
for prolonged periods of time (3 to 4 weeks), to single doses of 2000 to 6000 roentgens.
Dosage ranges for radioisotopes vary widely, and depend on the half-life of the isotope,
the strength and type of radiation emitted, and the uptake by the neoplastic cells. The
skilled artisan is directed to "Remington's Pharmaceutical Sciences" 15th Edition, chapter
33, in particular pages 624-652. Some variation in dosage will necessarily occur
depending on the condition of the subject being treated. The person responsible for
administration will, in any event, determine the appropriate dose for the individual
subject. Moreover, for human administration, preparations should meet sterility,
pyrogenicity, general safety and purity standards as required by FDA Office of Biologics
standards.
In preferred embodiments of the present invention, the regional delivery of the
nanodevice to patients with cancers is utilized to maximize the therapeutic effectiveness
of the delivered agent. Similarly, the chemo- or radiotherapy may be directed to
particular, affected region of the subjects body. Alternatively, systemic delivery of the
immunotherapeutic composition and/or the agent may be appropriate in certain
circumstances, for example, where extensive metastasis has occurred.
In addition to combining the nanodevice with chemo- and radiotherapies, it also is
contemplated that traditional gene therapies are used. For example, targeting of p53 or
pl6 mutations along with treatment of the nanodevices provides an improved anti-cancer
treatment. The present invention contemplates the co-treatment with other tumor-related
genes including, but not limited to, p21, Rb, APC, DCC, NF-I, NF-2, BCRA2, p16,
FHIT, WT-I, MEN-I, MEN-II, BRCA1, VHL, FCC, MCC, ras, myc, neu, raf. erb, src,
fins, jun, trk, ret, gsp, hst, bcl, and abl.
In vivo and ex vivo treatments are applied using the appropriate methods worked
out for the gene delivery of a particular construct for a particular subject. For example,
for viral vectors, one typically delivers 1 x 104, 1 x 10s, 1 x 106, 1 x 107, 1 x 108, 1 x
109, 1 x 1010, 1 x 1011 or 1 x 1012 infectious particles to the patient. Similar figures may
be extrapolated for liposomal or other non-viral formulations by comparing relative
uptake efficiencies.
An attractive feature of the present invention is that the therapeutic compositions
may be delivered to local sites in a patient by a medical device. Medical devices that are
suitable for use in the present invention include known devices for the localized delivery
of therapeutic agents. Such devices include, but are not limited to, catheters such as
injection catheters, balloon catheters, double balloon catheters, microporous balloon
catheters, channel balloon catheters, infusion catheters, perfusion catheters, etc., which
are, for example, coated with the therapeutic agents or through which the agents are
administered; needle injection devices such as hypodermic needles and needle injection
catheters; needleless injection devices such as jet injectors; coated stents, bifurcated
stmts, vascular grafts, stent grafts, etc.; and coated vaso-occlusive devices such as wire
coils.
Exemplary devices are described in U.S. Patent Nos. 5,935,114; 5,908,413;
5,792,105; 5,693,014; 5,674,192; 5,876,445; 5,913,894; 5,868,719; 5,851,228; 5,843,089;
5,800,519; 5,800,508; 5,800,391; 5,354,308; 5,755,722; 5,733,303; 5,866,561; 5,857,998;
5,843,003; and 5,933,145; the entire contents of which are incorporated herein by
reference. Exemplary stents that are commercially available and may be used in the
present application include the RADIUS (Scimed Life Systems, Inc.), the SYMPHONY
(Boston Scientific Corporation), the Wallstent (Schneider Inc.), the PRECEDENT II
(Boston Scientific Corporation) and the NIR (Medinol Inc.). Such devices are delivered
to and/or implanted at target locations within the body by known techniques.
XL Photodynamic Therapy
In some embodiments, the therapeutic complexes of the present invention
comprise a photodynamic compound and a targeting agent that is administred to a
patient. In some embodiments, the targeting agent is then allowed a period of time to
bind the 'target' cell (e.g. about 1 minute to 24 hours) resulting in the formation of a
target cell-target agent complex, In some embodiments, the therapeutic complexes
comprising the targeting agent and photodynamic compound are then illuminated (e.g.,
with a red laser, incandescent lamp, X-rays, or filtered sunlight). In some embodiments,
the light is aimed at the jugular vein or some other superficial blood or lymphatic vessel.
In some embodiments, the singlet oxygen and free radicals diffuse from the
photodynamic compound to the target cell (e.g. cancer cell or pathogen) causing its
destruction.
XII. Pharmaceutical Formulations
Where clinical applications are contemplated, in some embodiments of the present
invention, the nanodevices are prepared as part of a pharmaceutical composition in a
form appropriate for the intended application. Generally, this entails preparing
compositions that are essentially free of pyrogens, as well as other impurities that could
be harmful to humans or animals. However, in some embodiments of the present
invention, a straight dendrimer formulation may be administered using one or more of
the routes described herein.
In preferred embodiments, the nanodevices are used in conjunction with
appropriate salts and buffers to render delivery of the compositions in a stable manner to
allow for uptake by target cells. Buffers also are employed when the nanodevices are
introduced into a patient. Aqueous compositions comprise an effective amount of the
nanodevice to cells dispersed in a pharmaceutically acceptable carrier or aqueous
medium. Such compositions also are referred to as inocula. The phrase
"pharmaceutically or pharmacologically acceptable" refer to molecular entities and
compositions that do not produce adverse, allergic, or other untoward reactions when
administered to an animal or a human. As used herein, pharmaceutically acceptable
carrier" includes any and all solvents, dispersion media, coatings, antibacterial and
antifungal agents, isotonic and absorption delaying agents and the like. Except insofar as
any conventional media or agent is incompatible with the vectors or cells of the present
invention, its use in therapeutic compositions is contemplated. Supplementary active
ingredients may also be incorporated into the compositions.
In some embodiments of the present invention, the active compositions include
classic pharmaceutical preparations. Administration of these compositions according to
the present invention is via any common route so long as the target tissue is available via
that route. This includes oral., nasal., buccal., rectal., vaginal or topical. Alternatively,
administration may be by orthotopic, intradermal., subcutaneous, intramuscular,
intraperitoneal or intravenous injection.
The active nanodevices may also be administered parenterally or intraperitoneally
or intratumorally. Solutions of the active compounds as free base or pharmacologically
acceptable salts are prepared in water suitably mixed with, a surfactant, such as
hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid
polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of
storage and use, these preparations contain a preservative to prevent the growth of
microorganisms.
The pharmaceutical forms suitable for injectable use include sterile aqueous
solutions or dispersions and sterile powders for the extemporaneous preparation of sterile
injectable solutions or dispersions. The carrier may be a solvent or dispersion medium
containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol,
and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable
oils. The proper fluidity can be maintained, for example, by the use of a coating, such
as lecithin, by the maintenance of the required particle size in the case of dispersion and
by the use of surfactants. The prevention of the action of microorganisms can be
brought about by various antibacterial an antifungal agents, for example, parabens,
chlorobutanol, phenol, sorbic acid, thimerosal., and the like. In many cases, it may be
preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged
absorption of the injectable compositions can be brought about by the use in the
compositions of agents delaying absorption, for example, aluminum monostearate and
gelatin.
Sterile injectable solutions are prepared by incorporating the active compounds in
the required amount in the appropriate solvent with various of the other ingredients
enumerated above, as required, followed by filtered sterilization. Generally, dispersions
are prepared by incorporating the various sterilized active ingredients into a sterile
vehicle which contains the basic dispersion medium and the required other ingredients
from those enumerated above. In the case of sterile powders for the preparation of
sterile injectable solutions, the preferred methods of preparation are vacuum-drying and
freeze-drying techniques which yield a powder of the active ingredient plus any
additional desired ingredient from a previously sterile-filtered solution thereof.
Upon formulation, the dendrimer compositions are administered in a manner
compatible with the dosage formulation and in such amount as is therapeutically
effective. The formulations are easily administered in a variety of dosage forms such as
injectable solutions, drug release capsules and the tike. For parenteral administration in
an aqueous solution, for example, the solution is suitably buffered, if necessary, and the
liquid diluent first rendered isotonic with sufficient saline or glucose. These particular
aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and
intraperitoneal administration. For example, one dosage could be dissolved in 1 ml of
isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected
at the proposed site of infusion, (see for example, "Remington's Pharmaceutical
Sciences" 15th Edition, pages 1035-1038 and 1570-1580). In some embodiments of the
present invention, the active particles or agents are formulated within a therapeutic
mixture to comprise about 0.0001 to 1.0 milligrams, or about 0.001 to 0.1 milligrams, or
about 0.1 to 1.0 or even about 10 milligrams per dose or so. Multiple doses may be
administered.
Additional formulations that are suitable for other modes of administration include
vaginal suppositories and pessaries. A rectal pessary or suppository may also be used.
Suppositories are solid dosage forms of various weights and shapes, usually medicated,
for insertion into the rectum, vagina or the urethra. After insertion, suppositories soften,
melt or dissolve in the cavity fluids. In general., for suppositories, traditional binders and
carriers may include, for example, polyalkylene glycols or triglycerides; such
suppositories may be formed from mixtures containing the active ingredient in the range
of 0.5% to 10%, preferably l%-2%. Vaginal suppositories or pessaries are usually
globular or oviform and weighing about 5 g each. Vaginal medications are available in a
variety of physical forms, e.g., creams, gels or liquids, which depart from the classical
concept of suppositories. In addition, suppositories may be used in connection with
colon cancer. The nanodevices also may be formulated as inhalants for the treatment of
lung cancer and such like.
XIII. Method Of Treatment Or Prevention Of Cancer and Pathogenic
Diseases
In specific embodiments of the present invention methods and compositions are
provided for the treatment of tumors in cancer therapy. It is contemplated that the
present therapy can be employed in the treatment of any cancer for which a specific
signature has been identified or which can be targeted. Cell proliferative disorders, or
cancers, contemplated to be treatable with the methods of the present invention include
human sarcomas and carcinomas, including, but not limited to, fibrosarcoma,
myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma,
angiosarcoma, endotheliosarcoma, lymphangiosarcoma, Ewing's tumor,
lymphangioendotheliosarcoma, synovioma, mesothelioma, leiomyosarcoma,
rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer,
prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat
gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary
adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma,
renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma,
embryonal carcinoma, Wilms' tumor, cervical cancer, testicular tumor, lung carcinoma,
small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma,
medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma,
acoustic neuroma, oligodendroglioma, meningioma, melanoma, neuroblastoma,
retinoblastoma; leukemias, acute lymphocytic leukemia and acute myelocytic leukemia
(myeloblastic, promyelocytic, myelomonocytic, monocytic and erythroleukemia); chronic
leukemia (chronic myelocytic (granulocytic) leukemia and chronic lymphocytic
leukemia); and polycythemia vera, lymphoma (Hodgkin's disease and non-Hodgkin's
disease), multiple myeloma, Waldenstrom's macroglobulinemia, and heavy chain disease.
It is contemplated that the present therapy can be employed in the treatment of
any pathogenic disease for which a specific signature has been identified or which can be
targeted for a given pathogen. Examples of pathogens contemplated to be treatable with
the methods of the present invention include, but are not limited to, Legionella
peomophilia, Mycobacterium tuberculosis, Clostridium tetani, Hemophilus influenzae,
Neisseria gonorrhoeae, Treponema pallidum, Bacillus anthracis, Vibrio cholerae,
Borrelia burgdorferi, Cornebacterium diphtheria, Staphylococcus aureus, human
papilloma virus, human immunodeficiency virus, rubella virus, polio virus, and the like.
EXAMPLES
The following examples are provided to demonstrate and further illustrate certain
preferred embodiments of the present invention and are not to be construed as limiting
the scope thereof.
EXAMPLE 1
Quantitative MTT Biocompatibility/Cytotoxicity Assays
This Example describes quantitative MTT biocompatibility/cytotoxicity assays in
both mouse and rat primary fibroblasts to measure cytotoxicity of various individual
dendrimers and core-shell dendrimer molecules. In particular, the cytotoxicity of
PAMAM dendrimers (G5 and G7 generations), POPAM dendrimers (generations 2, 3,
and 4), and core-shell dendrimer molecules (i.e., POPAM 'core' dendrimer molecules
covalently linked to 2 or 3 PAMAM 'shell' dendrimers) were analyzed employing a
standard quantitative MTT assay (See Kuhlmann et al., 1998; Sladowski et al., 1993;
Wang et al., 1996; Watanabe et al., 1994).
Briefly, both the mouse and rat primary fibroblasts were cultured for 24 hours
with MTT (3-(4,5-dimethylthiazol-2-yi)-2 diphenyl tetrazolium bromide), and either
PAMAM dendrimers, POMAM dendrimers, or the core-shell dendrimer molecules. The
quantity of viable cells was then measured by absorbance at 540 nm in order to detect
the fonnazan product (purple) resulting from the cleavage of MTT (yellow) present only
in viable cells.
The results of these assays revealed a sharp distinction between the cytotoxicity of
the POPAM dendrimers and both the PAPAM dendrimers and the core-shell dendrimers
of the present invention. Specifically, the PAPAM dendrimers (G5 and G7 generations)
assayed produced no significant in vitro cytotoxicity at concentrations up to 40 jig/ml. In
contrast, the three types of POPAM dendrimers (generations 2, 3, and 4) induced
concentration-related cytotoxic effects with CL50 concentrations of 40, 12, and 12 μg/ml
respectively for murine fibroblasts, and 30, 9, and 9 ug/ml respectively for rat
fibroblasts. Interestingly, the core-shell dendrimer molecules did not share the
cytotoxicity problems of POPAM dendrimers as only concentrations of the core-shell
dendrimers higher than 30 μg/ml produced detectable toxicity, with only 5-10% of the
cells killed after 24 hours exposure to 40 ug/ml. These results demonstrate the favorable
biocompatability properties of the core-shell dendrimer molecules of the present
invention.
EXAMPLE 2
Construction of a Multifunctional Dendrimer Molecule
This example describes the construction of a multifunctional dendrimer molecule
with both targeting and signaling units. In particular, this example describes the
construction of a generation 5 (G5) PAMAM dendrimer conjugated to folic acid and
fluorescein where remaining amino surface groups on the dendrimer are 'capped' with
acetic anhydride or glycidol.
The schematic for production of the dendrimers if provided in Figure 8.
Conjugation of the G5 PAMAM dendrimers was carried out by reacting G5 PAMAM G5
with fluorescein isothiocyanate and N(Et)3. This fluorescein construct was then reacted
with folic acid and EDC. Reaction of the remaining amino surface groups of the product
with either acetic anhydride or glycidol resulted in their conversion to acetamido or
bis(2,3-hydroxypropyl)amino moieties, respectively. These biologically- and charge-
neutral "capping" groups gave the folate/fluorescein products and high aqueous solubility.
G5 PAMAM dendrimers were purified via ultrafiltration of pH-neutralized material in
1:1 DMSO/water.
EXAMPLE 3
In vitro Screening Assays
The toxicity and efficacy of the nanodevices of the present invention may be
assayed in vitro. In preferred embodiments, the nanodevices are tested in cell culture
models. For example, the efficacy of nanodevice for diagnosing, monitoring, and
treating breast cancer may assayed in breast cancer cell lines. For example, dendrimers
that target breast cancer cells are generated by conjugating ligands or antibodies that
specifically recognize receptors over-expressed by a particular breast cancer cell line.
For example, the SUM-52 cell line has an amplification of and over-expresses the FGFR-
2, c-MET, and NCAM-1 genes. The products of all of these genes are expressed to high
levels on the surface of SUM-52 cells and are not expressed to appreciable levels on
normal cells, or on other breast cancer cells. Libraries of dendrimers containing
candidate binding partners for any of these surface exposed factors are exposed to the
cells and candidate with specific and high binding affinity are identified. Similar assays
may be conducted with imaging components, therapeutic components, and the like. For
example, a library of dendrimers comprising different therapeutic agents are exposed to
the cell line. The ability of the agent to alter cell growth or kill the cell, while not
harming normal cells is screened. Ideally, such assays are conducted in multi-well plates
to allow the screening of large numbers of candidates simultaneouly or in a short time
period. In preferred embodiments, the screening assays are automated. For example,
screening for anti-cancer compounds that induce apoptosis can be automated by
providing a system for detecting the colorimetric changes induced by apoptosis (e.g.,
colorimetric changes caused by the imaging components of the present invention, as
described above).
Any number of cell lines may be used in the screening assays. For example, for
breast cancer, the cell lines SUM-190 and SUM-225 have an amplification of and
overexpress HER-2. Thus, antibodies, such as the humanized version of 4D5 (herceptin),
can be used to target dendrimers specifically to these cells. SUM-149, SUM-159, and
SUM-229 all over-express the EGFR. Thus, EGR, TGF-α, or amphiregulin are used to
target dendrimers to these cells. SUM-44 cells express HER-4 and thus are trageted
using heregulin-dendrimer conjugates. A variety of human mammary cell lines available
from ATCC may be used as controls including BT20, MCF7, UACC-893, and
UACC812. These cells differ in the expression of HER-2 and MUC1. Screening assays
may be performed in isolated cell populations and mixed cell populations.
EXAMPLE 4
Killing of Drug-resistant Cells
This example describes the killing of cisplatin resistant cell using cisplatin
conjugated to dendrimers. In these experiments, cell viability was assessed using the
tetrazolium-based colorimetric MTT assay (discussed in more detail below) (Mosmann, J.
Immunol. Meth., 65:55 [1983]). Human cell line 16N2 was grown in serum free, Ham's
F-12 medium supplemented with 5% BSA, insulin, and hydrocortisone. Cells were
seeded in 96-well microtitre plates at 1 x 104/well. After 24 hours, the medium was
changed and cisplatin (Stem Chemicals) or Dendrimer/Platin conjugates were added to
the wells. Cell viability was evaluated after 72 hours by MTT assay. The results are
shown in Figure 9. In Figure 9, drug concentration is expressed in platinum equivalents.
Results are expressed as a percentage of the dead cells with respect to control cells
grown in the absence of drug. Data represent mean +/- SEM (n=4). The Polymer 1 and
Polymer 2 samples are both generation 3.5 PAMAM dendrimers conjugated with
different content of platinum (E3.5-COONa:Pt with 19.25 and 20.26% of Pt,
respectively). The hydrogel compound is a generation 4 PAMAM dendrimer conjugated
with Pt (E4NH2:Pt gel containing 6.25% Pt).
Example 5
Methods for Preparing Oligonucleotide/Dendrimer Supramolecular Assemblies
As a first step toward controlling the oligonucleotides conjugation to the surface
primary amino groups of the dendrimers, partial and complete modification of amines
with acetic anhydride was performed. The number of the amino groups before and after
conjugation was determined by 1H NMR and potentiometric titration.
I) Complete acetamide capping of G7 and G5 PAMAM by N-acetylation
A dendrimer stock solution (10 wt% in MeOH; 0.05g, 0.43 μmol) was placed in a
25 mL round-bottom flask flushed with dry nitrogen, and 4 times molar excess of acetic
anhydride was added dropwise at 4°C. Triethylamine base (Aldrich) (0.23g; 2.3mmol)
was added to the reaction mixture with mild stirring at room temperature for 24 hours,
followed by addition of 1mL of MeOH (Aldrich, 99.8%) to dilute the mixture. This
solution was then allowed to react at room temperature for 24 hours. The product
solution was rotary evaporated to remove MeOH and transferred to 3.5k MWCO dialysis
tubing (Spectrum). It was then dialyzed against double distilled water (18.2MQ, MiliQ)
for 3 days, replacing the DI water 5 times during this period. After dialysis, the sample
was freeze-dried at -52°C for 24 hours to yield white power (57mg, 94%). The product
was analyzed by 1H-NMR.
2) Determination of actual number of terminal amino groups of G7 and G5.
The potentiometric titration of an aqueous solution of intact G7 and G5
dendrimers was performed using a Corning 420 pH meter with a Corning glass
combination electrode at 20°C. In brief, lyophilized G7 and G5 dendrimers were
dissolved respectively at 10 mL of 0.1N NaCl solution to prevent any electrostatic
interactions within the dendrimers caused by strong positive charges of amino group (See
e.g., Kabanov et al., Macromolecules 32:1904 [1999]). The dendrimers were fully
protonated by the addition of a 0. IN HC1 standard solution (Aldrich), then titrated with
0.1N NaOH standard solution at 3 min time intervals to achieve constancy to measure pH
values.
3) Partial acetamide capping of G7 and G5 PAMAM by stoichiometric
N-acetylation
Using the actual number of primary NH2 group on the surface of dendrimers, the
molar ratio of acetic anhydride to the amino groups was determined and reacted at same
method mentioned above. Each of core and shell dendrimer was expected to have 89%
and 90 % acetamide capping respectively by the addition of 89% of acetic anhydride to
the amino groups in terms of molar ratio. Table 1 summarized the stochiometry of the
partial acetylation for G7 and G5 PAMAM dendrimers.

4) Sequence design
The first 16 nucleotides of a 50-base oligomer served as a spacer and the last 34
serve as a recognition element for the complementary target sequence. Sequences were
selected such that on hybridization, recognition segments of the linkage could link the
core and shell dendrimers tightly together. In addition, the recognition segment was
designed to be cut by Sfil restriction enzyme so that any fragmentation pattern of this
tectodendrimer may be observed from use in vivo. The sequence analysis of the core
and shell oligonucleotides is as follows from the data of Vector NTI system.

This construct virtually eliminates hairpin formations in either
oligonucleotide and dramatically reduces the potential for core/core and shell/shell
hybridization. It also allows for more precise control of core/shell hybridization
using the differential of the Tm of the core/shell hybrid vs. the core/core or
shell/shell hybrids. This will further reduce the incidence of unwanted crosslinking
(gel formation). Furthermore, addition of extra nucleotides on the 3' end of and
between the restriction sites should improve the ability of Sfil to recognize and cut
the dsDNA recognition site during in vivo linker accessibility/degradation testing.
Thus, the theoretical formation of core/shell dimer was assumed as follows.
All publications and patents mentioned in the above specification are herein
incorporated by reference. Various modifications and variations of the described
method and system of the invention will be apparent to those skilled in the art
without departing from the scope and spirit of the invention. Although the
invention has been described in connection with specific preferred embodiments, it
should be understood that the invention as claimed should not be unduly limited to
such specific embodiments. Indeed, various modifications of the described modes
for carrying out the invention which are obvious to those skilled in biochemistry,
immunology, chemistry, molecular biology, the medical fields or related fields are
intended to be within the scope of the following claims.
WE CLAIM:
1. A composition comprising a dendrimer complex, wherein said first
dendrimer is covalently linked to said second dendrimer, said dendrimer
complex comprising first and second dendrimers, said first dendrimer
comprising a first agent and said second dendrimer comprising a second
agent, wherein said first agent is different than said second agent
2. The composition as claimed in claim 1, wherein said first and said second
agents are selected from the group consisting of therapeutic agents,
biological monitoring agents, biological imaging agents, targeting agents,
and agents capable of identifying a specific signature of cellular
abnormality.
3. The composition as claimed in claim 1, further comprising a third
dendrimer, wherein said third-dendrimer is covalently linked to said first
and said second dendrimers.
4. The composition as claimed in claim 3, further comprising a third agent
complexed with said third dendrimer.
5. The composition as claimed in claim 3, further comprising a fourth
dendrimer comprising a third agent, wherein said fourth dendrimer is
covalently linked to said third dendrimer,
6. The composition as claimed in claim 5, further comprising a fifth dendrimer
comprising a fourth agent, wherein said fifth dendrimer is complexed with
said third dendrimer.
7. The composition as claimed in claim 1, wherein said first agent is a
therapeutic agent and said second agent is a biological monitoring agent
8. The composition as claimed in claim 7, wherein said therapeutic agent is
selected from a chemotherapeutic agent, an anti-oncogenic agent, an anti-
vascularizing agent, and an expression construct comprising a nucleic
acid encoding a therapeutic protein.
9. The composition as claimed in claim 7, wherein said therapeutic agent is
protected with a protecting group selected from photo-labile, radio-labile,
and enzyme-labile protecting groups.
10. The composition as claimed in claim 8, wherein said chemotherapeutic
agent is selected from platinum complex, verapamil, podophyllotoxin,
carboplatin, procarbazine, mechlorethamine, cyclophosphamide,
camptothecin, ifosfamide, melphalan, chlorambucil, bisulfan, nitrosurea,
adriamycin, dactinomycin, daunorubicin, doxorubicin, bleomycin,
plicomycin, mitomycin, etoposide, tamoxifen, taxol, transplatinum, 5-
fluorouracil, vincristin, vinblastin, and methotrexate.
11. The composition as claimed in claim 8, wherein said anti-oncogenic agent
comprises an antisense nucleic acid.
12. The composition as claimed in claim 11, wherein said antisense nucleic
acid comprises a sequence complementary to an RNA of an oncogene.
13. The composition as claimed in claim 12, wherein said oncogene is
selected from abl, Bcl-2, Bcl-x1, erb, fms, gsp, hst, jun, myc, neu, raf, ras,
ret, src, or trk.
14. The composition as claimed in claim 8, wherein said nucleic acid encodes
a factor selected from tumor suppressor, cytokine, receptor, inducer of
apoptosis, or differentiating agent.
15. The composition as claimed in claim 14, wherein said tumor suppressor is
selected from BRCA1, BRCA2, C-CAM, p16, p21, p53, p73, Rb, and p27.
16. The composition as claimed in claim 14, wherein said cytokine is selected
from GMCSF, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11,
IL-12, IL-13, IL-14, IL-15, β-inteferon, γ-interferon, and TNF.
17. The composition as claimed in claim 14, wherein said receptor is selected
from CFTR, FGFR, estrogen receptor, IL-2 receptor, and VEGFR.
18. The composition as claimed in claim 14, wherein said inducer of apoptosis
is selected from AdE1B, Bad, Bak, Bax, Bid, Bik, Bim, Harakid, and ICE-
CED3 protease.
19. The composition as claimed in claim 2, wherein said biological monitoring
agent comprises an agent that measures an effect of a therapeutic agent.
20. The composition as claimed in claim 2, wherein said therapeutic agent
comprises a short-half life radioisotope.
21. The composition as claimed in claim 2, wherein said imaging agent
comprises a radioactive label selected from 14C, 36Cl, 57Co, 58Co, 51Cr, 125l,
131l, 111ln, 152Eu, 59Fe, 67Ga, 32P, 186Re, 35S, 75Se, Tc-99m, and 169Yb.
22. The composition as claimed in claim 19, wherein said monitoring agent is
capable of measuring the amount of apoptosis caused by said therapeutic
agent.
23. The composition as claimed in claim 2, wherein said targeting agent is
selected from antibody, receptor ligand, hormone, vitamin, and antigen.
24. The composition as claimed in claim 23, wherein said antibody is specific
for a disease specific antigen.
25. The composition as claimed in claim 24, wherein said disease specific
antigen comprises a tumor specific antigen.
26. The composition as claimed in claim 23, wherein said receptor ligand is
selected from a ligand for CFTR, EGFR, estrogen receptor, FGR2, folate
receptor, IL-2 receptor, glycoprotein, and VEGFR.,
27. The composition as claimed in claim 3, wherein said first and second
dendrimers comprise PAMAM dendrimers and wherein said third
dendrimer comprises a POPAM dendrimer.
28. The composition as claimed in claim 1, wherein said dendrimer complex
consists of said first dendrimer complexed with said second dendrimer.
29. The composition as claimed in claim 1, wherein said first dendrimer
comprises a first nucleic acid linker and said second dendrimer comprises
a second nucleic acid linker, wherein said first nucleic acid linker is
hybridized to said second nucleic acid linker.
30. The composition as claimed in claim 29, wherein a duplex formed from
hybridization of said first linker to said second linker comprises a cleavage
site.
31. The composition as claimed in claim 30, wherein said cleavage site
comprises a nuclease recognition site.
32. The composition as claimed in claim 31, wherein said nuclease
recognition site comprise a restriction endonuclease recognition site.

A composition comprising a dendrimer complex, wherein said first dendrimer is
covalently linked to said second dendrimer, said dendrimer complex comprising
first and second dendrimers, said first dendrimer comprising a first agent and
said second dendrimer comprising a second agent, wherein said first agent is
different than said second agent.

Documents:

IN-PCT-2002-1396-KOL-FORM-27.pdf

in-pct-2002-1396-kol-granted-abstract.pdf

in-pct-2002-1396-kol-granted-assignment.pdf

in-pct-2002-1396-kol-granted-claims.pdf

in-pct-2002-1396-kol-granted-correspondence.pdf

in-pct-2002-1396-kol-granted-description (complete).pdf

in-pct-2002-1396-kol-granted-drawings.pdf

in-pct-2002-1396-kol-granted-examination report.pdf

in-pct-2002-1396-kol-granted-form 1.pdf

in-pct-2002-1396-kol-granted-form 18.pdf

in-pct-2002-1396-kol-granted-form 2.pdf

in-pct-2002-1396-kol-granted-form 26.pdf

in-pct-2002-1396-kol-granted-form 3.pdf

in-pct-2002-1396-kol-granted-form 5.pdf

in-pct-2002-1396-kol-granted-reply to examination report.pdf

in-pct-2002-1396-kol-granted-specification.pdf

in-pct-2002-1396-kol-granted-translated copy of priority document.pdf


Patent Number 231426
Indian Patent Application Number IN/PCT/2002/1396/KOL
PG Journal Number 10/2009
Publication Date 06-Mar-2009
Grant Date 04-Mar-2009
Date of Filing 12-Nov-2002
Name of Patentee THE REGENTS OF THE UNIVERSITY OF MICHIGAN
Applicant Address 3003 SOUTH STATE STREET, ANN ARBOR, MI
Inventors:
# Inventor's Name Inventor's Address
1 TOMALIA DONALD, A. 426 VILLAGE GREEN BLVD, APT. 101, ANN ARBOR, MI 48105-3634
2 BAKER JAMES R, JR. 3997 HOLDEN DRIVE, ANN ARBOR, MI 48103
PCT International Classification Number A61K 47/48
PCT International Application Number PCT/US2001/15204
PCT International Filing date 2001-05-11
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 09/570,198 2000-05-12 U.S.A.