Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Jun 16.
Published in final edited form as: Bioconjug Chem. 2010 Jun 16;21(6):1014–1017. doi: 10.1021/bc1000802

Poly(amidoamine) Dendrimer Based MRI Contrast Agents Exhibiting Enhanced Relaxivities Derived via Metal Pre-ligation Techniques

Kido Nwe , L Henry Bryant Jr ξ, Martin W Brechbiel
PMCID: PMC2889128  NIHMSID: NIHMS206018  PMID: 20462240

This report presents the preparation and characterization of three [Gd-C-DOTA]−1-dendrimer assemblies by way of analysis, NMRD spectroscopy and Photon Correlation Spectroscopy (PCS). The metal-ligand chelates were preformed in alcohol media prior to conjugation to generation 4, 5 and 6 PAMAM dendrimers. The dendrimer-based agents were purified by Sephadex® G-25 column chromatography. The combustion analysis, SE-HPLC and UV-Vis data indicated chelate to dendrimer ratios of 28:1, 61:1 and 115:1 respectively. Molar relaxivity measured at pH 7.4, 22°C, and 3T (29.6, 49.8 and 89.1 mM−1s−1) indicated the viability of conjugates as MRI contrast agents. 1/T1 NMRD profiles were measured at 23°C and indicated that at 22 MHz the 1/T1 reached a plateau at 60, 85 and 140 mM−1s−1 for the generation 4, 5 and 6 dendrimer conjugates, respectively. The PCS data showed the respective size of 5.2, 6.5 and 7.8 nm for G-4, 5, and 6 conjugates.

During the development of MRI, contrast agents have been employed to induce additional contrast and increase the sensitivity of MRI scans.(1, 2) Increased usage of MRI, combined with the necessity for a contrast agent, prompts development of new efficient agents. Clinically used low molecular weight extracellular contrast agents, e.g. [Gd(DTPA)(H2O)]−2 (Magnevist®) suffer from rapid extravasation from blood vessels into interstitial space and fast decrease in concentration in blood vessels, combined with rapid whole body clearance.

In recent years, dendrimers have become an exciting class of polymeric platforms for assembly of multifunctional macromolecular nanomaterials undergoing pre-clinical development as diagnostics and therapeutic delivery vehicles. There have been reports indicating that dendrimers are applicable as carriers for site-specific delivery of drugs and that they do not alter the function of the molecules attached.(38) It has also been discovered that dendrimers can act as drugs themselves.(9, 10) In the MRI field, dendrimers have not only allowed molecules to retain their function, but have also greatly enhanced the signal to noise ratio of various imaging modalities; however, this application has never reached its full potential.

The use of Gd(III) chelates conjugated to high molecular weight carriers such as a PAMAM dendrimer prolongs intravascular retention and circulation time, slows down molecular rotation which results in a short relaxation time, and increases in relaxivity.(1113) Polyamidoamine (PAMAM) dendrimers of different generations conjugated to acyclic diethylenetriamine pentaacetic acid (DTPA) derivatives, e.g., 2-(p-isothiocyanatobenzyl)-6-methyl-diethylenetriaminepentaacetic acid (1B4M-DTPA), are versatile in MRI applications due to their mono-dispersed nature and available range of molecular sizes.(14, 15)

An acceptable contrast agent must be non-cytotoxic, must have high water solubility most preferably at physiological pH, and high relaxivity properties. The recent reports regarding nephrogenic systemic fibrosis (NSF) linked to the use of [Gd DTPA]−2 derived MR contrast agents provided our laboratory with an impetus to re-evaluate use of bifunctional DTPA agents for creation of macromolecular MR agents not only in regards to complex stability,(1620) but also in regards to their inherent extended in vivo residence time. [Gd(DOTA)]−1 (1,4,7,10-tetraazacyclododecane-N,N′,N″,N‴-tetraacetic acid) is known to be a better contrast agent in terms of complex stability and thus potentially less toxicity as compared to [Gd(DTPA)]−2.(21, 22) [Gd(DOTA)]−1, DOTAREM, has been extensively evaluated in patients with pre-existing renal diseases and to date without clinical side effects.(23) The higher relaxivity and kinetic stability of [Gd(DOTA)]−1 makes it an alternative contrast agent for MRI.(24) The formation and stability of the [Gd(DOTA)]−1 complex has also been thoroughly studied; dissociation of the metal ion was exceedingly slow even at low pH (24), on the order of days,(25) while its thermodynamic stability was comparable to [Gd(III)](DTPA).(26) [Gd(DOTA)]−1 was previously shown to be five orders of magnitude higher in in vitro stability as compared to [Gd(DTPA)]−2, which potentially translates to decreased in vivo toxicity.(21)

Herein, we report the preparation and characterization of three contrast agents with different generation (4, 5 and 6) of dendrimers. The C-DOTA (2-(4-nitrobenzyl)-1,4,7,10-tetraazacyclododecane-N,N′,N″,N‴-tetraacetic acid) ligand is first used to sequester Gd(III) (Scheme 1) and thereafter the formed metal complex is covalently attached to the terminal −NH2 groups of the dendrimer. We have recently reported that pre-metallation method to be significantly advantageous over conventional post-metallation methods, resulting in both a more directly characterizable product possessing an ~2-fold enhancement of molar relaxivity when using a bifunctional DTPA chelating agent while also decreasing the overall Gd(III) content by ~30%.(27) We have also reported the applicability of a C-DOTA derivative applied to complex Gd(III) as an agent of choice to replace the use of bifunctional DTPA agents employed to sequester Gd(III) as a component of macromolecular MR contrast agents.(28)

Scheme 1.

Scheme 1

Synthesis of dendrimer conjugates.

Table 1 summarizes the number of chelates per dendrimer obtained from analysis and SE-HPLC. For comparison, a UV-Vis spectrophotometric titration method was developed. A standard curve using compound 2 (Figure 1; Supporting Information), which represents a surrogate of the chelate conjugated to an amino surface on the dendrimer, was generated from this method and is shown in Figure 2 (Supporting Information). The results from assessing the number of chelates per dendrimer were collected in Table 1. Clearly, there is agreement between the two methods within experimental error indicating that spectrophotometric method also to be a viable method for the determination of the number of chelates conjugated to a dendrimer platform.

Table 1.

Number of chelates and molar relaxivity values measured for G4-(C-DOTA-Gd)28, G5-(C-DOTA-Gd)61 and G6-(C-DOTA-Gd)115.

Agent No. chelate (analysis) No. chelate (UV) r1 (mM−1s−1)
Magnevist --- --- 4.2
G4-(C-DOTA-Gd)28 28 26 29.6
G5-(C-DOTA-Gd)61 61 58 49.8
G6-(C-DOTA-Gd)115 113 107 89.1

A plot of the inverse of longitudinal relaxation time (1/T1) versus Gd(III) concentration for the three dendrimer conjugates and Magnevist is shown in Figure 3, while the relaxivity values obtained as a slope of this plot are summarized in Table 1. The data showed that the relaxivity of the three dendrimer based agents are 7–21 times higher than that of Magnevist. This indicates that G6-(C-DOTA-Gd)115 might be applicable in the submicromolar range andthus also presents the highest potential to eliminate concerns regarding Gd(III) toxicity.

Figure 3.

Figure 3

Molar relaxivity plots of G4-(C-DOTA-Gd)28 (■; 29.6 mM−1s−1), G5-(C-DOTA-Gd)61) (●; 49.8 mM−1s−1), G6-(C-DOTA-Gd)115) (▲; 89.1 mM−1s−1) and Magnevist (◆; 4.2 mM−1s−1).

The relaxivity values (30, 50 and 90 mM−1s−1) reported here produced by macromolecules measured at 3T field strength might seem high, but the reality is that these numbers are not uncommon. A relaxivity range of 139–237 mM−1s−1 at 2.4 T filed strength, which is close to our field strength, has been reported by Zhang et al in their contemporary work with fullerene.(29) The solutions prepared were based on the gadolinium content from the ICP report of the purified dendrimer conjugate. We reported the efficiency of this macromolecular purification method earlier,(27) which is also utilized by other researchers.(29)

In general, conjugation of compound 1 [Gd(C-DOTA))−1 (Scheme 1) to a higher generation dendrimer such as G6 would result in a more hydrophobic conjugate due to the replacement of exterior primary amines with the complexes. Hydrophobic interaction with solvent molecules is known to slow down the tumbling time (τR) which results in increase of the relaxivity.(3032) Based on the combustion analysis, SE-HPLC and UV-Vis data, the agent based on the G6 has the highest free amino sites (141), and thus it is expected to have the highest relaxivity. The experimental data (Table 1) support this position.

Consideration of the nuclear magnetic relaxation dispersion (NMRD) for a given contrast agent is essential since the field strength of clinical MRI instrumentation ranges from 2 to 180 MHz. The 1/T1 NMRD profiles of the G4, G5 and G6 dendrimer conjugates at 23°C are depicted in Figure 4, wherein the relaxivity for all three agents peaked at ~22 MHz. This behavior is similar to a previously reported measurement using G5, G7, G9 and G10 dendrimers in which the relaxivities peaked at ~22 MHz.(33, 34) Henry et. al. pointed out that the relaxivity of G7 conjugate is comparable to that of G6, which means G6 is the highest generation dendrimers where a significant increase in relaxivity can be observed based on the simple size and amine number of the dendrimers platform.(33, 34) However, whether that result will hold true for the agents prepared employing the pre-metallation method presented here remains to be determined in future studies.

Figure 4.

Figure 4

1/T1 NMRD profiles of 1 mM solution of prohance (▲), Magnevist® (●), G4-(C-DOTA-Gd)28(−), G5-(C-DOTA-Gd)61(◆), G6-(C-DOTA-Gd)115(■), as a function of frequencies at 23°C.

The size of conjugates determined here is based on 1 μM stock solution prepared in ambient water at pH = 6 to eliminate buffer interference (Table 2). As noted earlier, size measurement based on PCS is concentration dependent (35) and also buffer dependent.(36) As expected, the G6 conjugate has the largest size (Table 2) and thus has the highest impact on the relaxivity (Table 1). This is the first time that our laboratory has measured the size of PAMAM dendrimer conjugated to [Gd(C-DOTA)]−1. Relaxivity was observed to be linearly correlated to the size of chelated dendrimer conjugate; the larger the size, the greater the relaxivity (Figure 5; Supporting Information). This is in agreement with a previous report utilizing a similar [Gd(DO3A)]-derivative by Margerum et. al.(36), and other authors where subsequent increases in size and molecular weight results in increased relaxivity.(33, 34) This is also a major benefit from attaching a small chelate to a large macromolecule in which the associated tumbling time of the molecule increases leading to increases in relaxation enhancement.(37) Our NMRD data showed that the relaxivity of G=6 dendrimer chelate is 140 mM−1s−1 at 22 MHz, which is slightly lower than Gd(III)-glutamine synthetase (148 mM−1s−1) at 22.5MHz.(38, 39)

Table 2.

Values of size and ζ potential for free and conjugated dendrimers.

Generation Predicted Diameter* (nm) Measured Diameter (nm) ζ Potential
G4 4.0 4.2 0.1
G4 Conjugate 5.2 −60
G5 5.3 5.1 7.6
G5 Conjugate 6.5 −65
G6 6.7 6.4 17.4
G6 Conjugate 7.8 −69
*

Predicted values by Tomalia. (41)

The measurement of zeta potential (ζ) reveals the accumulation of ions at the particle surface.(35) The overall surface charge or ζ potential of the G6 agent is the highest due to the highest density of surface charge as expected. Also, high positive charge ζ potential is indicative of high electrostatic repulsion between particles and provides an energy barrier against aggregation.(35) Since the differences in ζ potential of the agents is not significant based on the data obtained, one can assume a likelihood of each agent to aggregate to be low. This will be advantageous for in vivo applications where aggregation could interfere with glomerulal filtration clearance through kidney, which would be the preferred route,(40) especially for larger G5 and G6 conjugates.

In summary, this study indicates that macromolecular-based MRI contrast agents prepared here are efficient and effective in modulating and relaxing water protons when compared to a single Gd(III) chelate. Our study also shows that appending the pre-formed Gd(III) complex to the dendrimer is far more convenient and significantly more advantageous in areas including species distribution, ease of characterization, stability, and solubility. As reported earlier, this pre-metallation strategy also produces a more hydrophobic agent that results in high relaxivity, and faster blood clearance rate in vivo.(27) The characterized data provide us with valuable information regarding the nature of the conjugates such as the size and the surface charge, and also enhance our understanding of how these parameters impact the relaxivity and to a certain degree what we can expect from these agents in vivo. These data strongly suggest that the G6-(C-DOTA-Gd)115 is a superior agent based on its high relaxivity which will also enable use of a much lower injected dose for all future studies thereby facilitating also translation into clinical trial evaluation.

Experimental

See supporting information

Supplementary Material

Supplementary Data

Acknowledgments

This research was supported by the Intramural Research Program of the National Institute of Health, National Cancer Institute, Center for Cancer Research and the United States Department of Health and Human Services.

Footnotes

Supporting Information Available

Detailed description of syntheses, molar relaxivity measurements, relaxometry, photon correlation spectroscopy (PCS) and zeta (ζ) potential, UV-Vis Spectroscopy and figures. This information is available free of charge via the Internet at http://pubs.acs.org/.

References

  • 1.Caravan P. Strategies for increasing the sensitivity of gadolinium based MRI contrast agents. Chem Soc Rev. 2006;35:512–523. doi: 10.1039/b510982p. [DOI] [PubMed] [Google Scholar]
  • 2.Villaraza AJL, Bumb A, Brechbiel MW. Macromolecules, Dendrimers, and Nanomaterials in Magnetic Resonance Imaging: The Interplay between Size, Function, and Pharmacokinetics. Chem Rev. 2010 doi: 10.1021/cr900232t. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Koyama Y, Talanov VS, Bernardo M, Hama Y, Regino CAS, Brechbiel MW, Choyke PL, Kobayashi H. A dendrimer-based nanosized contrast agent dual-labeled for magnetic resonance and optical fluorescence imaging to localize the sentinel lymph node in mice. J Magn Reson Imaging. 2007;25:866–871. doi: 10.1002/jmri.20852. [DOI] [PubMed] [Google Scholar]
  • 4.Lee CC, Gillies ER, Fox ME, Guillaudeu SJ, Frechet JMJ, Dy EE, Szoka FC. A single dose of doxorubicin-functionalized bow-tie dendrimer cures mice bearing C-26 colon carcinomas. Proc Nat Acad Sci. 2006;103:16649–16654. doi: 10.1073/pnas.0607705103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Gillies ER, Fréchet JMJ. Dendrimers and dendritic polymers in drug delivery. Drug Discov Today. 2005;10:35–43. doi: 10.1016/S1359-6446(04)03276-3. [DOI] [PubMed] [Google Scholar]
  • 6.Gurdag S, Khandare J, Stapels S, Matherly LH, Kannan RM. Activity of Dendrimer-Methotrexate Conjugates on Methotrexate-Sensitive and -Resistant Cell Lines. Bioconjugate Chem. 2006;17:275–283. doi: 10.1021/bc0501855. [DOI] [PubMed] [Google Scholar]
  • 7.Kukowska-Latallo JF, Candido KA, Cao Z, Nigavekar SS, Majoros IJ, Thomas TP, Balogh LP, Khan MK, Baker JR., Jr Nanoparticle Targeting of Anticancer Drug Improves Therapeutic Response in Animal Model of Human Epithelial Cancer. Cancer Res. 2005;65:5317–5324. doi: 10.1158/0008-5472.CAN-04-3921. [DOI] [PubMed] [Google Scholar]
  • 8.Kim Y, Hechler Ba, Gao Z-G, Gachet C, Jacobson KA. PEGylated Dendritic Unimolecular Micelles as Versatile Carriers for Ligands of G Protein-Coupled Receptors. Bioconjugate Chem. 2009;20:1888–1898. doi: 10.1021/bc9001689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Lee CC, MacKay JA, Frechet JMJ, Szoka FC. Nat Biotech. 2005;23:1517–1526. doi: 10.1038/nbt1171. [DOI] [PubMed] [Google Scholar]
  • 10.Menjoge AR, Kannan RM, Tomalia DA. Dendrimer-based drug and imaging conjugates: design considerations for nanomedical applications. Drug Discov Today. 2010;15:171–185. doi: 10.1016/j.drudis.2010.01.009. [DOI] [PubMed] [Google Scholar]
  • 11.Wang SJ, Brechbiel M, Wiener EC. Characteristics of a New MRI Contrast Agent Prepared From Polypropyleneimine Dendrimers, Generation 2. Invest Radiol. 2003;38:662–668. doi: 10.1097/01.rli.0000084887.47427.75. [DOI] [PubMed] [Google Scholar]
  • 12.Wiener E, Brechbiel MW, Brothers H, Magin RL, Gansow OA, Tomalia DA, Lauterbur PC. Dendrimer-based metal chelates: A new class of magnetic resonance imaging contrast agents. Magn Reson Med. 1994;31:1–8. doi: 10.1002/mrm.1910310102. [DOI] [PubMed] [Google Scholar]
  • 13.Tomalia DA, Reyna LA, Svenson S. Dendrimers as multi-purpose nanodevices for oncology drug delivery and diagnostic imaging. Biochem Soc Trans. 2007;035:61–67. doi: 10.1042/BST0350061. [DOI] [PubMed] [Google Scholar]
  • 14.Kobayashi H, Brechbiel MW. Dendrimer-based macromolecular MRI contrast agents:characteristic and application. Mol Imaging. 2003;2:1–10. doi: 10.1162/15353500200303100. [DOI] [PubMed] [Google Scholar]
  • 15.Sato N, Kobayashi H, Hiraga A, Saga T, Togashi K, Konishi J, Brechbiel MW. Pharmacokinetics and enhancement patterns of macromolecular MR contrast agents with various sizes of polyamidoamine dendrimer cores. Magn Reson Med. 2001;46:1169–1173. doi: 10.1002/mrm.1314. [DOI] [PubMed] [Google Scholar]
  • 16.Sieber MA, Lengsfeld P, Walter J, Schirmer H, Frenzel T, Siegmund F, Weinmann HJ, Pietsch H. Gadolinium-based contrast agents and their potential role in the pathogenesis of nephrogenic systemic fibrosis: The role of excess ligand. J Magn Reson Imaging. 2008;27:955–962. doi: 10.1002/jmri.21368. [DOI] [PubMed] [Google Scholar]
  • 17.Thakral C, Alhariri J, Abraham JL. Long-term retention of gadolinium in tissues from nephrogenic systemic fibrosis patient after multiple gadolinium-enhanced MRI scans: case report and implications. Contr Media Mol Imaging. 2007;2:199–205. doi: 10.1002/cmmi.146. [DOI] [PubMed] [Google Scholar]
  • 18.Stratta P, Canavese C, Aime S. Gadolinium-Enhanced Magnetic Resonance Imaging, Renal Failure and Nephrogenic Systemic Fibrosis/Nephrogenic Fibrosing Dermopathy. Curr Med Chem. 2008;15:1229–1235. doi: 10.2174/092986708784310396. [DOI] [PubMed] [Google Scholar]
  • 19.Port M, Idée J-M, Medina C, Robic C, Sabatou M, Corot C. Efficiency, thermodynamic and kinetic stability of marketed gadolinium chelates and their possible clinical consequences: a critical review. BioMetals. 2008;21:469–490. doi: 10.1007/s10534-008-9135-x. [DOI] [PubMed] [Google Scholar]
  • 20.Abraham JL, Chandra S, Thakral C, Abraham JM. SIMS imaging of gadolinium isotopes in tissue from Nephrogenic Systemic Fibrosis patients: Release of free Gd from magnetic resonance imaging (MRI) contrast agents. Appl Surf Sci. 2008;255:1181–1184. [Google Scholar]
  • 21.Bousquet JC, Saini S, Stark DD, Hahn PF, Nigam M, Wittenberg J, Ferrucci JT., Jr Gd-DOTA: characterization of a new paramagnetic complex. Radiol. 1988;166:693–698. doi: 10.1148/radiology.166.3.3340763. [DOI] [PubMed] [Google Scholar]
  • 22.Allard M, Doucet D, Kien P, Bonnemain B, Caille JM. Experimental study of DOTA-Gadolinium pharmacokinetics and pharmacologic properties. Invest Radiol. 1988;23(suppl 1):S271–S274. doi: 10.1097/00004424-198809001-00059. [DOI] [PubMed] [Google Scholar]
  • 23.Deray G, Bellin MF, Baumelou B, Rey JP, Boulechfar H, Grellet J, Jacobs C. Renal Tolerance of the Paramagnetic Contrast Medium Gd-DOTA in Patients with Chronic Renal Failure. Am J Nephrol. 1990;10:522–523. doi: 10.1159/000168180. [DOI] [PubMed] [Google Scholar]
  • 24.Magerstädt M, Gansow OA, Brechbiel MW, Colcher D, Baltzer L, Knop RH, Girton ME, Naegele M. Gd(DOTA): An alternative to Gd(DTPA) as a T1,2 relaxation agent for NMR imaging or spectroscopy. Magn Reson Med. 1986;3:808–812. doi: 10.1002/mrm.1910030517. [DOI] [PubMed] [Google Scholar]
  • 25.Wang X, Jin T, Comblin V, Lopez-Mut A, Merciny E, Desreux JF. A kinetic investigation of the lanthanide DOTA chelates. Stability and rates of formation and of dissociation of a macrocyclic gadolinium(III) polyaza polycarboxylic MRI contrast agent. Inorg Chem. 1992;31:1095–1099. [Google Scholar]
  • 26.Benazeth S, Purans J, Chalbot MC, Nguyen-van-Duong MK, Nicolas L, Keller F, Gaudemer A. Temperature and pH Dependence XAFS Study of Gd(DOTA)- and Gd(DTPA)2- Complexes: Solid State and Solution Structures. Inorg Chem. 1998;37:3667–3674. doi: 10.1021/ic9707321. [DOI] [PubMed] [Google Scholar]
  • 27.Nwe K, Xu H, Regino CAS, Bernardo M, Ileva L, Riffle L, Wong KJ, Brechbiel MW. A New Approach in the Preparation of Dendrimer-Based Bifunctional Diethylenetriaminepentaacetic Acid MR Contrast Agent Derivatives. Bioconjugate Chem. 2009;20:1412–1418. doi: 10.1021/bc900057z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Nwe K, Bernardo M, Regino CAS, Williams M, Brechbiel MW. Comparison of magnetic resonane imaging (MRI) properties between derivatized diethylenetriamine-N,N′,N′,N″, N″-pentaacetic acid (DTPA) and 1,4,7,10-tetraazacyclododecane-N,N′,N″,N‴-tetraacetic acid (DOTA) gadolinium- dendrimer conjugates. 2010 Manuscript Submitted. [Google Scholar]
  • 29.Zhang J, Fatouros PP, Shu C, Reid J, Owens LS, Cai T, Gibson HW, Long GL, Corwin FD, Chen Z-J, Dorn HC. High Relaxivity Trimetallic Nitride (Gd3N) Metallofullerene MRI Contrast Agents with Optimized Functionality. Bioconjugate Chemistry. doi: 10.1021/bc900375n. ASAP. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Wang SJ, Brechbiel MW, Weiner EC. Characteristics of a new MRI contrast agent prepared from polypropyleneimine dendrimers, generation 2. Invest Radiol. 2003;38:662–668. doi: 10.1097/01.rli.0000084887.47427.75. [DOI] [PubMed] [Google Scholar]
  • 31.Kellar KE, Henrichs PM, Hollister R, Koenig SH, Eck J, Wei D. High relaxivity linear Gd(DTPA)-polymer conjugates: the role of hydrophobic interactions. Magn Reson Med. 1997;38:712–716. doi: 10.1002/mrm.1910380506. [DOI] [PubMed] [Google Scholar]
  • 32.Zech SG, Eldredge HB, Lowe MP, Caravan P. Protein binding to lanthanide(III) complexes can reduce the water exchange rate at the lanthanide. Inorg Chem. 2007;46:3576–3584. doi: 10.1021/ic070011u. [DOI] [PubMed] [Google Scholar]
  • 33.Bryant LH, Jr, Martin WB, Wu C, Bulte JWM, Herynek V, Frank JA. Synthesis and relaxometry of high-generation (G = 5, 7, 9, and 10) PAMAM dendrimer-DOTA-gadolinium chelates. J Magn Reson Imaging. 1999;9:348–352. doi: 10.1002/(sici)1522-2586(199902)9:2<348::aid-jmri30>3.0.co;2-j. [DOI] [PubMed] [Google Scholar]
  • 34.Weiner EC, Brechbiel MW. Dendrimer-based metal chelates: a new class of magnetic resonance imaging contrast agents. Magn Reson Med. 1994;31:1–8. doi: 10.1002/mrm.1910310102. [DOI] [PubMed] [Google Scholar]
  • 35.Suvarna S, Espinasse B, Qi R, Lubica R, Poncz M, Cines DB, Wiesner MR, Arepally GM. Determinants of PF4/heparin immunogenicity. Blood. 2007;110:4253–4260. doi: 10.1182/blood-2007-08-105098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Margerum LD, Campion BK, Koo M, Shargill N, Lai J-J, Marumoto A, Christian Sontum P. Gadolinium(III) DO3A macrocycles and polyethylene glycol coupled to dendrimers Effect of molecular weight on physical and biological properties of macromolecular magnetic resonance imaging contrast agents. J Alloys Compd. 1997;249:185–190. [Google Scholar]
  • 37.Koenig SH, Brown RD., III Relaxation of solvent protons by paramagnetic ions and its dependence on magnetic field and chemical environment: implications for NMR imaging. Magn Reson Med. 1984;1:478–495. doi: 10.1002/mrm.1910010407. [DOI] [PubMed] [Google Scholar]
  • 38.Eads CD, Mulqueen P, Horrocks WD, Villafranca JJ. Comparative study of glutamine synthetase bound lanthanide(III) ions using NMR relaxation and lanthanide(III) luminescence techniques. Biochemistry. 1985;24:1221–1226. doi: 10.1021/bi00326a025. [DOI] [PubMed] [Google Scholar]
  • 39.Lauffer RB. Paramagnetic metal complexes as water proton relaxation agents for NMR imaging: theory and design. Chemical Reviews. 1987;87:901–927. [Google Scholar]
  • 40.Vexler VS, Clément O, Schmitt-Willich H, Brasch RC. Effect of varying the molecular weight of the MR contrast agent Gd-DTPA-polylysine on blood pharmacokinetics and enhancement patterns. J Magn Reson Imaging. 1994;4:381–388. doi: 10.1002/jmri.1880040325. [DOI] [PubMed] [Google Scholar]
  • 41.Donald AT, Adel MN, William AG., III Starburst Dendrimers: Molecular-Level Control of Size, Shape, Surface Chemistry, Topology, and Flexibility from Atoms to Macroscopic Matter. Angew Chem Int Ed Engl. 1990;29:138–175. [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary Data

RESOURCES