Poly(amidoamine)

Last updated

Poly(amidoamine), or PAMAM, is a class of dendrimer which is made of repetitively branched subunits of amide and amine functionality. PAMAM dendrimers, sometimes referred to by the trade name Starburst, have been extensively studied since their synthesis in 1985, [1] and represent the most well-characterized dendrimer family as well as the first to be commercialized. [2] Like other dendrimers, PAMAMs have a sphere-like shape overall, and are typified by an internal molecular architecture consisting of tree-like branching, with each outward 'layer', or generation, containing exponentially more branching points. This branched architecture distinguishes PAMAMs and other dendrimers from traditional polymers, as it allows for low polydispersity and a high level of structural control during synthesis, and gives rise to a large number of surface sites relative to the total molecular volume. Moreover, PAMAM dendrimers exhibit greater biocompatibility than other dendrimer families, perhaps due to the combination of surface amines and interior amide bonds; these bonding motifs are highly reminiscent of innate biological chemistry and endow PAMAM dendrimers with properties similar to that of globular proteins. [2] The relative ease/low cost of synthesis of PAMAM dendrimers (especially relative to similarly-sized biological molecules such as proteins and antibodies), along with their biocompatibility, structural control, and functionalizability, have made PAMAMs viable candidates for application in drug development, biochemistry, and nanotechnology. [2] [3] [4]

Contents

Synthesis

Divergent synthesis

A general scheme for the divergent synthesis of PAMAM dendrimers, with ethylene diamine as a core initiator. The scheme is color-coded by generation number, with the red ethylene diamine core serving as initiator core, orange as Generation 0 and orange/green as Generations 1, respectively. The scheme shown is currently the most widely adopted approach in commercial syntheses of PAMAM. PAMAM Divergent Synthesis Color-Coded.png
A general scheme for the divergent synthesis of PAMAM dendrimers, with ethylene diamine as a core initiator. The scheme is color-coded by generation number, with the red ethylene diamine core serving as initiator core, orange as Generation 0 and orange/green as Generations 1, respectively. The scheme shown is currently the most widely adopted approach in commercial syntheses of PAMAM.

Divergent synthesis refers to the sequential "growth" of a dendrimer layer by layer, starting with a core "initiator" molecule which contains functional groups capable of acting as active sites in the initial reaction. Each subsequent reaction in the series increases the number of available surface groups exponentially. Core molecules which give rise to PAMAM dendrimers can vary, but the most basic initiators are ammonia and ethylene diamine. [6] Outward growth of PAMAM dendrimers is accomplished by alternating between two reactions:

  1. Michael addition of the amino-terminated surface onto methyl acrylate, resulting in an ester-terminated outer layer, and
  2. Coupling with ethylene diamine to achieve a new amino-terminated surface.

Each round of reactions forms a new "generation", and PAMAM dendrimers are often classified by generation number; the common shorthand for this classification is "GX" or "GX PAMAM", where X is a number referring to the generation number. The first full cycle of Michael addition followed by coupling with ethylene diamine forms Generation 0 PAMAM, with subsequent Michael additions giving rise to "half" generations, and subsequent amide coupling giving rise to "full" (integer) generations.

With divergent synthesis of dendrimers, it is extremely important to allow each reaction to proceed to completion; any defects caused by incomplete reaction or intramolecular coupling of new surface amines with unreacted methyl ester surface groups could cause "trailing" generations, stunting further growth for certain branches. These impurities are difficult to remove when using the divergent synthetic approach because the molecular weight, physical size, and chemical properties of the defective dendrimers are very similar in nature to the desired product. As generation number increases, it becomes more difficult to produce pure products in a timely fashion due to steric constraints. As a result, synthesis of higher-generation PAMAM dendrimers can take months.

Convergent synthesis

Convergent synthesis of a dendrimer begins with what will eventually become the surface of the dendrimer and proceeds inward. The convergent synthetic approach makes use of orthogonal protecting groups (two protecting groups whose deprotection conditions will not remove one another); this is an additional consideration not present when using a divergent approach. The figure below depicts a general scheme for a convergent synthetic approach.

A generalized scheme outlining the use of orthogonal protecting groups for convergent synthesis of PAMAM dendrimers. PAMAM Convergent Synthesis.png
A generalized scheme outlining the use of orthogonal protecting groups for convergent synthesis of PAMAM dendrimers.

Convergent synthesis as shown above begins with the dendritic subunit composed of reactive "focal group" A and branched group B (B can be multiply branched in the most generalized scenario, but PAMAMs only split once at each branching point). First, A is orthogonally protected and set aside for further reactions. B is also orthogonally protected, leaving the unprotected A on this molecule to couple with each of the unprotected B groups from the initial compound. This results in a new higher-generation species that is protected on both A and B. Selective deprotection of A yields a new molecule which can again be coupled onto the original monomer, thus forming another new generation. This process can then be repeated to form more and more layers.

Toxicity

in vitro

It has been established that cationic macromolecules in general destabilize the cell membrane, which can lead to lysis and cell death. [9] The common conclusion present in current work echoes this observation: increasing dendrimer molecular weight and surface charge (both being generation-dependent) increases their cytotoxic behavior. [10] [11] [12] [13] [14] [15]

Initial studies on PAMAM toxicity showed that PAMAM was less toxic (in some cases, much less so) than related dendrimers, [16] exhibiting minimal cytotoxicity across multiple toxicity screens, including tests of metabolic activity (MTT assay), cell breakdown (LDH assay), and nucleus morphology (DAPI staining). [10] However, in other cell lines, the MTT assay and several other assays revealed some cytotoxicity. [12] [13] These disparate observations could be due to differences in sensitivity of the various cell lines used in each study to PAMAM; although cytotoxicity for PAMAM varies among cell lines, they remain less toxic than other dendrimer families overall.

More recently, a series of studies by Mukherjee et al. [13] [14] [15] have shed some light on the mechanism of PAMAM cytotoxicity, providing evidence that the dendrimers break free of their encapsulating membrane (endosome) after being absorbed by the cell, causing harm to the cell's mitochondria and eventually leading to cell death. Further elucidation of the mechanism of PAMAM cytotoxicity would help resolve the dispute as to precisely how toxic the dendrimers are.

In relation to neuronal toxicity, fourth generation PAMAM has been shown to break down calcium transients, altering neurotransmitter vesicle dynamics and synaptic transmission. All of the above can be prevented by replacing the surface amines with folate or polyethylene glycol. [17]

It has also been shown that PAMAM dendrimers cause rupturing of red blood cells, or hemolysis. [12] Thus, if PAMAM dendrimers are to be considered in biological applications that involve dendrimers or dendrimer complexes traveling through the bloodstream, the concentration and generation number of unmodified PAMAM in the bloodstream should be taken into account.

in vivo

To date, few in-depth studies on the in vivo behavior of PAMAM dendrimers have been carried out. This could be in part due to the diverse behavior of PAMAMs depending on surface modification (see below), which make characterization of their in vivo properties largely case-dependent. Nonetheless, the fate and transport of unmodified PAMAM dendrimers is an important case study as any biological applications could involve unmodified PAMAM as a metabolic byproduct. In the only major systematic study of in vivo PAMAM behavior, injections of high levels of bare PAMAMs over extended periods of time in mice showed no evidence of toxicity up through G5 PAMAM, and for G3-G7 PAMAM, low immunogenicity was observed. [11] These systemic-level observations seem to align with the observation that PAMAM dendrimers are not extremely cytotoxic overall; however, more in-depth studies of the pharmacokinetics and biodistribution of PAMAM are required before a move toward in vivo applications can be made.

Surface modification

One unique property of dendrimers such as PAMAM is the high density of surface functional groups, which allow many alterations to be made to the surface of each dendrimer molecule. In putative PAMAM dendrimers, the surface is rife with primary amines, with higher generations expressing exponentially greater densities of amino groups. Although the potential to attach many things to each dendrimer is one of their greatest advantages, the presence of highly localized positive charges can be toxic to cells. Surface modification via attachment of acetyl [18] and lauroyl [19] groups help mask these positive charges, attenuating cytotoxicity and increasing permeability to cells. Thus, these types of modifications are especially beneficial for biological applications. Secondary and tertiary amino surface groups are also found to be less toxic than primary amino surface groups, [10] suggesting it is charge shielding which has major bearing on cytotoxicity and not some secondary effect from a particular functional group. Furthermore, other studies point to a delicate balance in charge which must be achieved to obtain minimal cytotoxicity. Hydrophobic interactions can also cause cell lysis, and PAMAM dendrimers whose surfaces are saturated with nonpolar modifications such as lipids or polyethylene glycol (PEG) suffer from higher cytotoxicity than their partially substituted analogues. [19] PAMAM dendrimers with nonpolar internal components have also been shown to induce hemolysis. [12]

Applications

Applications involving dendrimers in general take advantage of either stuffing cargo into the interior of the dendrimer (sometimes referred to as the "dendritic box"), or attaching cargo onto the dendrimer surface. PAMAM dendrimer applications have generally focused on surface modification, taking advantage of both electrostatic and covalent methods for binding cargo. Currently, major areas of study using PAMAM dendrimers and their functionalized derivatives involve drug delivery and gene delivery.

Drug delivery

Since PAMAM dendrimers have shown penetration capability to a wide range of cell lines, simple PAMAM-drug complexes would affect a broad spectrum of cells upon introduction to a living system. Thus, additional targeting ligands are required for the selective penetration of cell types. For example, PAMAM derivatized with folic acid is preferentially taken up by cancer cells, which are known to overexpress the folate receptor on their surfaces. Attaching additional treatment methods along with the folic acid, such as boron isotopes, [20] cisplatin, [21] and methotrexate have proven quite effective. [22] In the future, as synthetic control over dendrimer surface chemistry becomes more robust, PAMAM and other dendrimer families may rise to prominence alongside other major approaches to targeted cancer therapy.

In a study of folic acid functionalized PAMAM, methotrexate was combined either as an inclusion complex within the dendrimer or as a covalent surface attachment. In the case of the inclusion complex, the drug was released from the dendrimer interior almost immediately when subjected to biological conditions and acted similarly to the free drug. The surface attachment approach yielded stable, soluble complexes which were able to selectively target cancer cells and did not prematurely release their cargo. [22] Drug release in the case of the inclusion complex could be explained by the protonation of surface and interior amines under biological conditions, leading to unpacking of the dendrimer conformation and consequent release of the inner cargo. A similar phenomenon was observed with complexes of PAMAM and cisplatin. [23]

PAMAM dendrimers have also demonstrated intrinsic drug properties. One quite notable example is the ability for PAMAM dendrimers to remove prion protein aggregates, [24] the deadly protein aggregates responsible for bovine spongiform encephalopathy ("mad cow disease") and Creutzfeldt–Jakob disease in humans. The solubilization of prions is attributed to the polycationic and dendrimeric nature of the PAMAMs, with higher generation (>G3) dendrimers being the most efficient; hydroxy-terminated PAMAMs as well as linear polymers showed little to no effect. Since there are no other known compounds capable of dissolving prions which have already aggregated, PAMAM dendrimers have offered a bit of reprieve in the study of such fatal diseases, and may offer additional insight into the mechanism of prion formation.

Gene therapy

Surface amine residues on PAMAM dendrimers bind to the phosphate backbone of nucleic acids through charged interactions (right, inset). Typically, G6-7 PAMAM dendrimers are used for gene transfection; these dendrimers are typically 6-10nm in length (spanning ~20-30 base pairs) and have a molecular mass of 30-50kDa. PAMAM DNA Complexation.png
Surface amine residues on PAMAM dendrimers bind to the phosphate backbone of nucleic acids through charged interactions (right, inset). Typically, G6-7 PAMAM dendrimers are used for gene transfection; these dendrimers are typically 6-10nm in length (spanning ~20-30 base pairs) and have a molecular mass of 30-50kDa.

The discovery that mediating positive charge on PAMAM dendrimer surfaces decreases their cytotoxicity has interesting implications for DNA transfection applications. Because the cell membrane has a negatively charged exterior, and the DNA phosphate backbone is also negatively charged, the transfection of free DNA is not very efficient simply due to charge repulsion. However, it would be reasonable to expect charged interactions between the anionic phosphate backbone of DNA and the amino-terminated surface groups of PAMAM dendrimers, which are positively ionized under physiological conditions. This could result in a PAMAM-DNA complex, which would make DNA transfection more efficient due to neutralization of the charges on both elements, while the cytotoxicity of the PAMAM dendrimer would also be reduced. Indeed, several reports have confirmed PAMAM dendrimers as effective DNA transfection agents. [16] [26] [27] [28]

When the charge balance between DNA phosphates and PAMAM surface amines is slightly positive, the maximum transfection efficiency is obtained; [23] this finding supports the idea that the complex binds to the cell surface via charge interactions. A striking observation is that "activation" of PAMAM by partial degradation via hydrolysis improves transfection efficiency by 2-3 orders of magnitude, [23] providing further evidence supporting the existence of an electrostatically coupled complex. The fragmentation of some branches of the dendrimer is thought to loosen up the overall structure (fewer amide bonds and space constraints), which would theoretically result in better contact between the dendrimer and DNA substrate because the dendrimer is not forced into a rigid spherical conformation due to sterics. This in turn results in more compact DNA complexes which are more easily endocytosed. After endocytosis, the complexes are subjected to the acidic conditions of the cellular endosome. The PAMAM dendrimers act as a buffer in this environment, soaking up the excess protons with multitudes of amine residues, leading to the inhibition of pH-dependent endosomal nuclease activity and thus protecting the cargo DNA. The tertiary amines on the interior of the dendrimer can also participate in the buffering activity, causing the molecule to puff up; additionally, as the PAMAMs take on more and more positive charge, fewer of them are required for the optimal PAMAM-DNA interaction, and free dendrimers are released from the complex. Dendrimer release and swelling can eventually lyse the endosome, resulting in release of the cargo DNA. The activated PAMAM dendrimers have less spatial barrier to interior amine protonation, which is thought to be a major source of their advantage over non-activated PAMAM. [25]

PAMAM dendrimers can be "activated" for gene transfer applications via hydrolysis accelerated by heat, a process which can be thought of as similar to shearing bushes. During this process, amide bonds are broken and replaced with carboxyl groups (see inset), causing some branches of the dendrimer to fall off. The overall molecular mass of the dendrimer is reduced by 20-25%, and the result is a more flexible dendrimer with transfection efficiencies improved by 2-3 orders of magnitude. Activation of PAMAM via hydrolysis.png
PAMAM dendrimers can be "activated" for gene transfer applications via hydrolysis accelerated by heat, a process which can be thought of as similar to shearing bushes. During this process, amide bonds are broken and replaced with carboxyl groups (see inset), causing some branches of the dendrimer to fall off. The overall molecular mass of the dendrimer is reduced by 20-25%, and the result is a more flexible dendrimer with transfection efficiencies improved by 2-3 orders of magnitude.

In the context of existing approaches to gene transfer, PAMAM dendrimers hold a strong position relative to major classical technologies such as electroporation, microinjection, and viral methods. Electroporation, which involves pulsing electricity through cells to create holes in the membrane through which DNA can enter, has obvious cytotoxic effects and is not appropriate for in vivo applications. On the other hand, microinjection, the use of fine needles to physically inject genetic material into the cell nucleus, offers more control but is a high-skill, meticulous task in which a relatively low number of cells can be transfected. Although viral vectors can offer highly specific, high-efficiency transfection, the generation of such viruses is costly and time-consuming; furthermore, the inherent viral nature of the gene transfer often triggers an immune response, thus limiting in vivo applications. In fact, many modern transfection technologies are based on artificially assembled liposomes (both liposomes and PAMAMs are positively charged macromolecules). [25] Since PAMAM dendrimers and their complexes with DNA exhibit low cytotoxicity, higher transfection efficiencies than liposome-based methods, and are effective across a broad range of cell lines, [16] they have taken an important place in modern gene therapy methodologies. The biotechnology company Qiagen currently offers two DNA transfection product lines (SuperFect and PolyFect) based on activated PAMAM dendrimer technology.

Much work lies ahead before activated PAMAM dendrimers can be used as in vivo gene therapy agents. Although the dendrimers have proved to be highly efficient and non-toxic in vitro, the stability, behavior, and transport of the transfection complex in biological systems has yet to be characterized and optimized. As with drug delivery applications, specific targeting of the transfection complex is ideal and must be explored as well.

See also

Related Research Articles

Transfection is the process of deliberately introducing naked or purified nucleic acids into eukaryotic cells. It may also refer to other methods and cell types, although other terms are often preferred: "transformation" is typically used to describe non-viral DNA transfer in bacteria and non-animal eukaryotic cells, including plant cells. In animal cells, transfection is the preferred term as transformation is also used to refer to progression to a cancerous state (carcinogenesis) in these cells. Transduction is often used to describe virus-mediated gene transfer into eukaryotic cells.

<span class="mw-page-title-main">Colloidal gold</span> Suspension of gold nanoparticles in a liquid

Colloidal gold is a sol or colloidal suspension of nanoparticles of gold in a fluid, usually water. The colloid is coloured usually either wine red or blue-purple . Due to their optical, electronic, and molecular-recognition properties, gold nanoparticles are the subject of substantial research, with many potential or promised applications in a wide variety of areas, including electron microscopy, electronics, nanotechnology, materials science, and biomedicine.

<span class="mw-page-title-main">Dendrimer</span> Highly ordered, branched polymeric molecule

Dendrimers are highly ordered, branched polymeric molecules. Synonymous terms for dendrimer include arborols and cascade molecules. Typically, dendrimers are symmetric about the core, and often adopt a spherical three-dimensional morphology. The word dendron is also encountered frequently. A dendron usually contains a single chemically addressable group called the focal point or core. The difference between dendrons and dendrimers is illustrated in the top figure, but the terms are typically encountered interchangeably.

<span class="mw-page-title-main">Maleimide</span> Chemical compound

Maleimide is a chemical compound with the formula H2C2(CO)2NH (see diagram). This unsaturated imide is an important building block in organic synthesis. The name is a contraction of maleic acid and imide, the -C(O)NHC(O)- functional group. Maleimides also describes a class of derivatives of the parent maleimide where the NH group is replaced with alkyl or aryl groups such as a methyl or phenyl, respectively. The substituent can also be a small molecule (such as biotin, a fluorescent dye, an oligosaccharide, or a nucleic acid), a reactive group, or a synthetic polymer such as polyethylene glycol. Human hemoglobin chemically modified with maleimide-polyethylene glycol is a blood substitute called MP4.

<span class="mw-page-title-main">Nanochemistry</span> Combination of chemistry and nanoscience

Nanochemistry is an emerging sub-discipline of the chemical and material sciences that deals with the development of new methods for creating nanoscale materials. The term "nanochemistry" was first used by Ozin in 1992 as 'the uses of chemical synthesis to reproducibly afford nanomaterials from the atom "up", contrary to the nanoengineering and nanophysics approach that operates from the bulk "down"'. Nanochemistry focuses on solid-state chemistry that emphasizes synthesis of building blocks that are dependent on size, surface, shape, and defect properties, rather than the actual production of matter. Atomic and molecular properties mainly deal with the degrees of freedom of atoms in the periodic table. However, nanochemistry introduced other degrees of freedom that controls material's behaviors by transformation into solutions. Nanoscale objects exhibit novel material properties, largely as a consequence of their finite small size. Several chemical modifications on nanometer-scaled structures approve size dependent effects.

Cell-penetrating peptides (CPPs) are short peptides that facilitate cellular intake and uptake of molecules ranging from nanosize particles to small chemical compounds to large fragments of DNA. The "cargo" is associated with the peptides either through chemical linkage via covalent bonds or through non-covalent interactions.

Magnetofection is a transfection method that uses magnetic fields to concentrate particles containing vectors to target cells in the body. Magnetofection has been adapted to a variety of vectors, including nucleic acids, non-viral transfection systems, and viruses. This method offers advantages such as high transfection efficiency and biocompatibility which are balanced with limitations.

<span class="mw-page-title-main">Computational gene</span>

A computational gene is a molecular automaton consisting of a structural part and a functional part; and its design is such that it might work in a cellular environment.

<span class="mw-page-title-main">Polyethylenimine</span> Chemical compound

Polyethylenimine (PEI) or polyaziridine is a polymer with repeating units composed of the amine group and two carbon aliphatic CH2CH2 spacers. Linear polyethyleneimines contain all secondary amines, in contrast to branched PEIs which contain primary, secondary and tertiary amino groups. Totally branched, dendrimeric forms were also reported. PEI is produced on an industrial scale and finds many applications usually derived from its polycationic character.

A nanogel is a polymer-based, crosslinked hydrogel particle on the sub-micron scale. These complex networks of polymers present a unique opportunity in the field of drug delivery at the intersection of nanoparticles and hydrogel synthesis. Nanogels can be natural, synthetic, or a combination of the two and have a high degree of tunability in terms of their size, shape, surface functionalization, and degradation mechanisms. Given these inherent characteristics in addition to their biocompatibility and capacity to encapsulate small drugs and molecules, nanogels are a promising strategy to treat disease and dysfunction by serving as delivery vehicles capable of navigating across challenging physiological barriers within the body. 

Carbon nanotubes (CNTs) are very prevalent in today's world of medical research and are being highly researched in the fields of efficient drug delivery and biosensing methods for disease treatment and health monitoring. Carbon nanotube technology has shown to have the potential to alter drug delivery and biosensing methods for the better, and thus, carbon nanotubes have recently garnered interest in the field of medicine.

Dendrosomes are novel vesicular, spherical, supramolecular entities wherein the dendrimer–nucleic acid complex is encapsulated within a lipophilic shell. They possess negligible hemolytic toxicity and higher transfection efficiency, and they are better tolerated in vivo than are dendrimers. The word " Dendrosome" came from the Greek word "Dendron" meaning tree and " some" means vesicles. Thus dendrosomes are vesicular structures composed of dendrimers.

<span class="mw-page-title-main">Nanoparticle–biomolecule conjugate</span> Tailored macromolecule with covalently-bonded bio-active substances targeting specific tissues

A nanoparticle–biomolecule conjugate is a nanoparticle with biomolecules attached to its surface. Nanoparticles are minuscule particles, typically measured in nanometers (nm), that are used in nanobiotechnology to explore the functions of biomolecules. Properties of the ultrafine particles are characterized by the components on their surfaces more so than larger structures, such as cells, due to large surface area-to-volume ratios. Large surface area-to-volume-ratios of nanoparticles optimize the potential for interactions with biomolecules.

<span class="mw-page-title-main">Vectors in gene therapy</span>

Gene therapy utilizes the delivery of DNA into cells, which can be accomplished by several methods, summarized below. The two major classes of methods are those that use recombinant viruses and those that use naked DNA or DNA complexes.

Dendrimer-encapsulated nanoparticles (DENs) are nanoparticles ranging from 1.5 to 10 nm that are synthesized by a template approach using dendrimers. Monometallic, bimetallic and semiconductor nanoparticles have been synthesized using this method. The primary use for DEN's is as a catalyst due to their extremely high surface area to volume ratio. Advantages that DENs have over other nanoparticles is that they are monodisperse and easy to make.

<span class="mw-page-title-main">DNA-functionalized quantum dots</span>

DNA-functionalization of quantum dots is the attachment of strands of DNA to the surface of a quantum dot. Although quantum dots with cadmium (Cd) have some cytotoxic release, researchers have functionalized quantum dots for biocompatibility and bound them to DNA in order to combine the advantages of both materials. Quantum dots are commonly used for imaging biological systems in vitro and in vivo in animal studies due to their excellent optical properties when excited by light, while DNA has numerous bioengineering applications, including: genetic engineering, self-assembling nanostructures, protein binding, and biomarkers. The ability to visualize the chemical and biological processes of DNA allows feedback to optimize and learn about these small scale behaviors.

<span class="mw-page-title-main">Gold nanoparticles in chemotherapy</span> Drug delivery technique using gold nanoparticles as vectors

Gold nanoparticles in chemotherapy and radiotherapy is the use of colloidal gold in therapeutic treatments, often for cancer or arthritis. Gold nanoparticle technology shows promise in the advancement of cancer treatments. Some of the properties that gold nanoparticles possess, such as small size, non-toxicity and non-immunogenicity make these molecules useful candidates for targeted drug delivery systems. With tumor-targeting delivery vectors becoming smaller, the ability to by-pass the natural barriers and obstacles of the body becomes more probable. To increase specificity and likelihood of drug delivery, tumor specific ligands may be grafted onto the particles along with the chemotherapeutic drug molecules, to allow these molecules to circulate throughout the tumor without being redistributed into the body.

Nanoparticle drug delivery systems are engineered technologies that use nanoparticles for the targeted delivery and controlled release of therapeutic agents. The modern form of a drug delivery system should minimize side-effects and reduce both dosage and dosage frequency. Recently, nanoparticles have aroused attention due to their potential application for effective drug delivery.

Hamid Ghandehari is an Iranian-American drug delivery research scientist, and a professor in the Departments of Pharmaceutics and Pharmaceutical Chemistry and Biomedical Engineering at the University of Utah. His research is focused in recombinant polymers for drug and gene delivery, nanotoxicology of dendritic and inorganic constructs, water-soluble polymers for targeted delivery and poly(amidoamine) dendrimers for oral delivery.

<span class="mw-page-title-main">Intracellular delivery</span> Scientific research area

Intracellular delivery is the process of introducing external materials into living cells. Materials that are delivered into cells include nucleic acids, proteins, peptides, impermeable small molecules, synthetic nanomaterials, organelles, and micron-scale tracers, devices and objects. Such molecules and materials can be used to investigate cellular behavior, engineer cell operations or correct a pathological function.

References

  1. Tomalia DA, Baker H, Dewald J, Hall M, Kallos G, Martin S, et al. (1985). "A New Class of Polymers: Starburst-Dendritic Macromolecules". Polymer Journal. 17: 117–32. doi: 10.1295/polymj.17.117 .
  2. 1 2 3 Esfand R, Tomalia DA (April 2001). "Poly(amidoamine) (PAMAM) dendrimers: from biomimicry to drug delivery and biomedical applications". Drug Discovery Today. 6 (8): 427–436. doi:10.1016/S1359-6446(01)01757-3. PMID   11301287. Dendritech Inc "Dendritech". Weihai CY Dendrimer Technology Co., Ltd "CY Dendrimer".[ permanent dead link ]
  3. Lee CC, MacKay JA, Fréchet JM, Szoka FC (December 2005). "Designing dendrimers for biological applications". Nature Biotechnology. 23 (12): 1517–26. doi:10.1038/nbt1171. PMID   16333296. S2CID   2140256.
  4. Tomalia DA, Reyna LA, Svenson S (February 2007). "Dendrimers as multi-purpose nanodevices for oncology drug delivery and diagnostic imaging". Biochemical Society Transactions. 35 (Pt 1): 61–7. doi:10.1042/BST0350061. PMID   17233602.
  5. "Dendritech".
  6. Tomalia DA, Baker H, Dewald J, Hall M, Kallos G, Martin S, et al. (1986). "Dendritic macromolecules: Synthesis of starburst dendrimers". Macromolecules. 19 (9): 2466–8. Bibcode:1986MaMol..19.2466T. doi:10.1021/ma00163a029.
  7. Lee JW, Kim JH, Kim BK, Kim JH, Shin WS, Jin SH (2006). "Convergent synthesis of PAMAM dendrimers using click chemistry of azide-functionalized PAMAM dendrons". Tetrahedron. 62 (39): 9193–200. doi:10.1016/j.tet.2006.07.030.
  8. Hecht S (2003). "Functionalizing the interior of dendrimers: Synthetic challenges and applications". Journal of Polymer Science Part A: Polymer Chemistry. 41 (8): 1047–58. Bibcode:2003JPoSA..41.1047H. doi: 10.1002/pola.10643 .
  9. Rittner K, Benavente A, Bompard-Sorlet A, Heitz F, Divita G, Brasseur R, Jacobs E (February 2002). "New basic membrane-destabilizing peptides for plasmid-based gene delivery in vitro and in vivo". Molecular Therapy. 5 (2): 104–14. doi: 10.1006/mthe.2002.0523 . PMID   11829517.
  10. 1 2 3 Fischer D, Li Y, Ahlemeyer B, Krieglstein J, Kissel T (March 2003). "In vitro cytotoxicity testing of polycations: influence of polymer structure on cell viability and hemolysis". Biomaterials. 24 (7): 1121–31. doi:10.1016/S0142-9612(02)00445-3. PMID   12527253.
  11. 1 2 Roberts JC, Bhalgat MK, Zera RT (January 1996). "Preliminary biological evaluation of polyamidoamine (PAMAM) Starburst dendrimers". Journal of Biomedical Materials Research. 30 (1): 53–65. doi:10.1002/(SICI)1097-4636(199601)30:1<53::AID-JBM8>3.0.CO;2-Q. PMID   8788106.
  12. 1 2 3 4 Malik N, Wiwattanapatapee R, Klopsch R, Lorenz K, Frey H, Weener JW, et al. (March 2000). "Dendrimers: relationship between structure and biocompatibility in vitro, and preliminary studies on the biodistribution of 125I-labelled polyamidoamine dendrimers in vivo". Journal of Controlled Release. 65 (1–2): 133–48. doi:10.1016/S0168-3659(99)00246-1. PMID   10699277.
  13. 1 2 3 Mukherjee SP, Davoren M, Byrne HJ (February 2010). "In vitro mammalian cytotoxicological study of PAMAM dendrimers - towards quantitative structure activity relationships". Toxicology in Vitro. 24 (1): 169–77. doi:10.1016/j.tiv.2009.09.014. PMID   19778601.
  14. 1 2 Mukherjee SP, Lyng FM, Garcia A, Davoren M, Byrne HJ (November 2010). "Mechanistic studies of in vitro cytotoxicity of poly(amidoamine) dendrimers in mammalian cells". Toxicology and Applied Pharmacology. 248 (3): 259–68. doi:10.1016/j.taap.2010.08.016. PMID   20736030.
  15. 1 2 Mukherjee SP, Byrne HJ (February 2013). "Polyamidoamine dendrimer nanoparticle cytotoxicity, oxidative stress, caspase activation and inflammatory response: experimental observation and numerical simulation". Nanomedicine. 9 (2): 202–11. doi: 10.1016/j.nano.2012.05.002 . PMID   22633897.
  16. 1 2 3 Haensler J, Szoka FC (1993). "Polyamidoamine cascade polymers mediate efficient transfection of cells in culture". Bioconjugate Chemistry. 4 (5): 372–9. doi:10.1021/bc00023a012. PMID   8274523.
  17. Vidal F, Vásquez P, Cayumán FR, Díaz C, Fuentealba J, Aguayo LG, et al. (December 2017). "Prevention of Synaptic Alterations and Neurotoxic Effects of PAMAM Dendrimers by Surface Functionalization". Nanomaterials. 8 (1): 7. doi: 10.3390/nano8010007 . PMC   5791094 . PMID   29295581.
  18. Kolhatkar RB, Kitchens KM, Swaan PW, Ghandehari H (2007). "Surface acetylation of polyamidoamine (PAMAM) dendrimers decreases cytotoxicity while maintaining membrane permeability". Bioconjugate Chemistry. 18 (6): 2054–60. doi:10.1021/bc0603889. PMID   17960872.
  19. 1 2 Jevprasesphant R, Penny J, Attwood D, McKeown NB, D'Emanuele A (October 2003). "Engineering of dendrimer surfaces to enhance transepithelial transport and reduce cytotoxicity". Pharmaceutical Research. 20 (10): 1543–50. doi:10.1023/A:1026166729873. PMID   14620505. S2CID   30840885.
  20. Shukla S, Wu G, Chatterjee M, Yang W, Sekido M, Diop LA, et al. (2003). "Synthesis and biological evaluation of folate receptor-targeted boronated PAMAM dendrimers as potential agents for neutron capture therapy". Bioconjugate Chemistry. 14 (1): 158–67. doi:10.1021/bc025586o. PMID   12526705.
  21. Malik N, Evagorou EG, Duncan R (September 1999). "Dendrimer-platinate: a novel approach to cancer chemotherapy". Anti-Cancer Drugs. 10 (8): 767–76. doi:10.1097/00001813-199909000-00010. PMID   10573209.
  22. 1 2 Patri AK, Kukowska-Latallo JF, Baker JR (December 2005). "Targeted drug delivery with dendrimers: comparison of the release kinetics of covalently conjugated drug and non-covalent drug inclusion complex". Advanced Drug Delivery Reviews. 57 (15): 2203–14. doi:10.1016/j.addr.2005.09.014. PMID   16290254.
  23. 1 2 3 Boas U, Heegaard PM (January 2004). "Dendrimers in drug research". Chemical Society Reviews. 33 (1): 43–63. doi:10.1039/b309043b. PMID   14737508.
  24. Supattapone S, Nguyen HO, Cohen FE, Prusiner SB, Scott MR (December 1999). "Elimination of prions by branched polyamines and implications for therapeutics". Proceedings of the National Academy of Sciences of the United States of America. 96 (25): 14529–34. Bibcode:1999PNAS...9614529S. doi: 10.1073/pnas.96.25.14529 . JSTOR   121438. PMC   24470 . PMID   10588739.
  25. 1 2 3 4 Dennig J, Duncan E (May 2002). "Gene transfer into eukaryotic cells using activated polyamidoamine dendrimers". Journal of Biotechnology. 90 (3–4): 339–47. doi:10.1016/S1389-0352(01)00066-6. PMID   12071232.
  26. Bielinska A, Kukowska-Latallo JF, Johnson J, Tomalia DA, Baker JR (June 1996). "Regulation of in vitro gene expression using antisense oligonucleotides or antisense expression plasmids transfected using starburst PAMAM dendrimers". Nucleic Acids Research. 24 (11): 2176–82. doi:10.1093/nar/24.11.2176. PMC   145901 . PMID   8668551.
  27. Kukowska-Latallo JF, Bielinska AU, Johnson J, Spindler R, Tomalia DA, Baker JR (May 1996). "Efficient transfer of genetic material into mammalian cells using Starburst polyamidoamine dendrimers". Proceedings of the National Academy of Sciences of the United States of America. 93 (10): 4897–902. Bibcode:1996PNAS...93.4897K. doi: 10.1073/pnas.93.10.4897 . PMC   39376 . PMID   8643500.
  28. Bielinska AU, Chen C, Johnson J, Baker JR (1999). "DNA complexing with polyamidoamine dendrimers: implications for transfection". Bioconjugate Chemistry. 10 (5): 843–50. doi:10.1021/bc990036k. PMID   10502352.

Bibliography