Abstract

Various glutamate urea ligands have displayed high affinities to prostate specific membrane antigen (PSMA), which is highly overexpressed in prostate and other cancer sites. The multivalent versions of small PSMA-targeted molecules are known to be even more efficiently bound to the receptor. Here, we employ a well-known urea-based ligand, 2-[3-(1,3-dicarboxypropyl)-ureido] pentanedioic acid (DUPA) and triazine dendrimers in order to study the effect of molecular size on multivalent targeting in prostate cancer. The synthetic route starts with the preparation of a dichlorotriazine bearing DUPA in 67% overall yield over five steps. This dichlorotriazine reacts with G1, G3, and G5 triazine dendrimers bearing a 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) group for 64Cu-labeling at the core to afford poly(monochlorotriazine) intermediates. Addition of 4-aminomethylpiperidine (4-AMP) and the following deprotection produce the target compounds, G1-(DUPA)4, G3-(DUPA)16, and G5-(DUPA)64. These targets include 4/16/64 DUPA groups on the surface and a DOTA group at the core, respectively. In vitro cell assay using PC3-PIP (PSMA positive) and PC3-FLU (PSMA negative) cells reveals that G1-(DUPA)4 has the highest PC3-PIP to PC3-FLU uptake ratio (10-fold) through the PSMA-mediated specific uptake. While G5-(DUPA)64 displayed approximately 12 times higher binding affinity (IC50 23.6 nM) to PC3-PIP cells than G1-(DUPA)4 (IC50 282.3 nM) as evaluated in a competitive binding assay, the G5 dendrimer also showed high non-specific binding to PC3-FLU cells. In vivo uptake of the 64Cu-labeled dendrimers was also evaluated in severe combined inmmunodeficient (SCID) mice bearing PC3-PIP and PC3-FLU xenografts on each shoulder, respectively. Interestingly, quantitative imaging analysis of positron emission tomograph (PET) displayed the lowest tumor uptake in PC3-PIP cells for the midsize dendrimer G3-(DUPA)16 (19.4 kDa) (0.66 ± 0.15%ID/g at 1 h. p.i., 0.64 ± 0.11%ID/g at 4 h. p.i., and 0.67 ± 0.08%ID/g at 24 h. p.i.). Through the specific binding of G1-(DUPA)4 to PSMA, the smallest dendrimer (5.1 kDa) demonstrated the highest PC3-PIP to muscle and PC3-PIP to PC3-FLU uptake ratios (17.7 ± 5.5 and 6.7 ± 3.0 at 4 h p.i., respectively). In addition, the enhanced permeability and retention (EPR) effect appeared to be an overwhelming factor for tumor uptake of the largest dendrimer G5-(DUPA)64 as the uptake was at a similar level irrelevant to the PSMA expression.

Highlights

  • Targeted delivery is one of the key elements in the design of new drugs as it can improve their safety and efficacy by reducing the nonspecific uptake-associated toxicity, increasing the maximum tolerated dose (MTD), and enhancing the cellular uptake in specific disease sites [1,2]

  • The enhanced permeability and retention (EPR) effect appeared to be an overwhelming factor for tumor uptake of the largest dendrimer G5-(DUPA)64 as the uptake was at a similar level irrelevant to the prostate specific membrane antigen (PSMA) expression

  • Various cell surface markers have been evaluated for diagnosis and treatment of patients suffering from prostate cancer, including prostate specific antigen (PSA) [6], prostatic acid phosphatase (PAP) [7], prostate stem cell antigen (PSCA) [8], and prostate specific membrane antigen (PSMA) [9,10]

Read more

Summary

Introduction

Targeted delivery is one of the key elements in the design of new drugs as it can improve their safety and efficacy by reducing the nonspecific uptake-associated toxicity, increasing the maximum tolerated dose (MTD), and enhancing the cellular uptake in specific disease sites [1,2]. A generation 5 PAMAM dendrimer-methotrexate conjugate bearing multiple copies of glutamate urea (GLA) was evaluated with PSMA positive LNCaP cells in vitro and displayed elevated inhibition of the NAALADase activity when compared with free GLA at equimolar concentrations [41]. A recombinant protein of human serum albumin (HSA, 67 kDa) was localized up to three times more in a tumor over time when fused with a tumor targeting agent, amino-terminal fragment (ATF) [45] To date, it has been largely unclear how the interplay of the molecular size, multivalency, and receptor-binding affinity of nanomedicines affects their in vivo kinetics and targeted uptake. Tumor uptake of the dendrimers in a mouse model of prostate cancer is evaluated as therapeutic moiety, and a DOTA for 64Cu-labeling.

Procedures
Pomper of John Hopkins
Serum Stability Assay
Competition Assay
Cell Uptake and Internalization
Synthesis
The starts with preparation of protected diamine diamine
Radiolabeling and Serum Stability
64 Cu-labeled dendrimers and the internalization
16. A certain ofextent non-specific cellular uptake can be observed highly
Conclusions
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call