Designing ParaCEST Agents (BASIC, Responsive)
Marty Pagel1

1Cancer Systems Imaging, MD Anderson Cancer Center, houston, TX, United States

Synopsis

This presentation will review Paramagnetic Chemical Exchange Saturation Transfer (ParaCEST) MRI contrast agents. These agents should be thoroughly characterized with regard to their dependence on saturation time, saturation power, concentration, pH and temperature. Responsive ParaCEST agents can detect or measure enzyme activity, metabolites, metal ions, pH, redox state, temperature, and light. Some ParaCEST agents can also exhibit T2-Exchange relaxation. The intermediate exchange rate of a T2ex agent does not affect the T1 relaxivity of the agent. Therefore, the T2/T1 ratio of a T2ex agent can be employed to detect a biomarker.

Paramagnetic Chemical Exchange Saturation Transfer (ParaCEST) MRI contrast agents were first reported in 2001 and 2002 [1-3]. These agents can have a large saturation frequency (chemical shift of a labile proton), which allows for a chemical exchange rate as fast as 10,000 Hz that can generate a CEST signal with srong amplitude [4]. A ParaCEST agent typically consists of a macrocyclic chelate that holds a lanthanide ion, such as Eu(III), Tm(III), or Yb(III) [5]. Linear chelates and other lathanide ions have also been used as ParaCEST agents. To avoid the potential toxicity of lanthanide ions, chelates with iron, nickel and chromium have also been developed as paraCEST agents [6]. The labile proton that undergoes chemical exchange can be an amide, amine, or hydroxyl group of a ligand on the agent, or can be a water molecule that is bound to the metal of the agent.

ParaCEST agents should be thoroughly characterized before they are used in experiments. The effect of saturation time on the CEST signal amplitude can be assessed using the non-linear QUEST method [7] or the linear RL-QUEST method [8]. The effect of saturation power on the CEST signal amplitude can also be evaluated, and the average chemical exchange rate of the agent can be measured by using the non-linear QUESP method [7], a linear Omega Plot [9], or the HW-QUESP method [10]. The concentration dependence of the CEST signal amplitude under steady state saturation conditions can be evaluated using the linear HW-conc method [11]. In addition, the effects of pH on the CEST signal amplitude should be evaluated from 6.0 to 8.0 in small increments to assess a range of physiological conditions [12]. The effects of temperature should be evaluated between 22-52 °C [13]. Other conditions such as the effects of salt, proteins, cell homogenates or plasma can also be evaluated.

ParaCEST agents that are responsive to a biomarker comprise 20% (24/117) of all responsive MRI contrast agents reported as of 2014, demonstrating the creativity in the development of these agents [14,15]. ParaCEST agents have been developed that respond to enzyme activity [16]; metabolites such as sugars, lactate, phosphates, and nitric oxide [17]; metals such as zinc, calcium, and copper; pH [18]; redox state; temperature; and light. Some ParaCEST agents have been developed that have a biomarker-responsive CEST signal and an unresponsive CEST signal [13], or two CEST signals that respond differently to the biomarker [12]. A ratio of these two CEST signals can be used to detect or measure the biomarker in a manner that is independent of agent’s concentration, the T1 relaxation time constant of the sample, and B1 inhomogeneity of the saturation period [12,13]. This ratiometric approach facilitates in vivo studies with responsive ParaCEST agents [19,20].

Some ParaCEST agents can also exhibit T2-Exchange relaxation [21]. A physical exchange of protons between a T2ex agent and water causes the MR frequency of the exchanging proton to take a weighted average value between the frequency of the agent and the frequency of water. Because chemical exchange is stochastic, each proton experiences a different weighted average for its MR frequency. These different MR frequencies result in the decay of net coherent magnetization, which manifests as T2 relaxation. T2ex agents typically have chemical exchange rates of 100,000-1,000,000 Hz that are intermediate between the exchange rates of 10,000,000-1,000,000,000 Hz for T1 agents and 10-10,000 Hz for CEST agents [4]. This intermediate rate greatly improves detection sensitivity of T2ex agents relative to CEST agents, but T2ex agents have less sensitivity than T1 agents. The chemical exchange rate of bound water in a T2ex agent can be estimated by measuring the linewidth of 17O NMR spectra over a range of temperatures [22]. The chemical shift of bound water can be estimated based on the chemical shift of a proton on the macrocyclic chelate [22]. Finally, the temperature dependence of the T2ex relaxation should be evaluated to further confirm that the chemical exchange rate of the agent is in the intermediate rate regime [23].

T2ex relaxation can cause the CEST signal amplitude to decrease, so that paraCEST agents should be designed to avoid chemical exchange rates that create T2ex [24]. This apparent disadvantage can be exploited as an advantage to create responsive contrast agents that change from slow to intermediate chemical exchange rates (or vice-versa), which causes the agent to change from a CEST agent to a T2ex agent (or vice-versa) [23]. Furthermore, the intermediate exchange rate of a T2ex agent does not affect the T1 relaxivity of the agent that depends on faster chemical exchange rates. Therefore, a change in intermediate chemical exchange rate by a biomarker can alter T2 relaxation without altering T1 relaxation [23]. The T2/T1 ratio can then be employed to detect the biomarker without a dependence on the concentration of the agent. This approach has been used to develop a responsive T2ex agent that detects nitric oxide [23].

Acknowledgements

No acknowledgement found.

References

1. Woods M, Woessner DW, Sherry AD. Paramagnetic lanthanide complexes as PARACEST agents for medical imaging. Chem Soc Rev 2006;35(6):500–511.

2. Zhang S, Winter P, Wu K, Sherry AD. A novel europium(III)-based MRI contrast agent. J Am Chem Soc 2001;123(7):1517–1518.

3. Aime S, Delli Castelli D, Terreno E. Novel pH-reporter MRI contrast agents. Angew Chemie Int Ed 2002;41:4334-4336.

4. Wu Y, Sherry AD. The importance of water exchange rates in the design of responsive agents for MRI. Curr Opin Chem Biol 2013;17:167-174.

5. Aime S, Barge A, Delli Castelli D, Fedeli F, Mortillaro A, Nielsen FU, Terreno E. Paramagnetic lanthanide(III) complexes as pH-sensitive chemical exchange saturation transfer (CEST) contrast agents for MRI applications. Magn Reson Med 2002;47(4):639–648.

6. Tsitovich PB, Burns PJ, McKay AM, Morrow JR. Redox-activated MRI contrast agents based on lanthanide and transition metal ions. J Inorg Biochem 2014;133:143-154.

7. McMahon MT, Gilad AA, Zhou J, Sun PZ, Bulte JW, van Zijl PC. Quantifying exchange rates in chemical exchange saturation transfer agents using the saturation time and saturation power dependencies of the magnetization transfer effect on the magnetic resonance imaging signal (QUEST and QUESP): pH calibration for poly-L-lysine and a starburst dendrimer. Magn Reson Med 2006;55:836-847.

8. Randtke EA, Chen LQ, Pagel MD. The reciprocal linear QUEST analysis method facilitates the measurements of chemical exchange rates with CEST MRI. Contrast Media Molec Imaging 2014;9:252–258.

9. Dixon WT, Ren J, Lubag AJ, Ratnakar J, Vinogradov E, Hancu I, Lenkinski RE, Sherry AD. A concentration-independent method to measure exchange rates in PARACEST agents. Magn Reson Med 2010;63:625-632.

10. Randtke EA, Chen LQ, Corrales LR, Pagel MD. The Hanes-Woolf Linear QUESP method improves the measurements of fast chemical exchange rates with CEST MRI. Magn Reson Med 2014;71:1603-1612.

11. Ali MM, Liu G, Shah T, Flask CA, Pagel MD. Using two chemical exchange saturation transfer magnetic resonance imaging contrast agents for molecular imaging studies. Acc Chem Res 2009;42:915-924.

12. Sheth VR, Liu G, Li Y, Pagel MD. Improved pH measurements with a single PARACEST MRI contrast agent. Contrast Media Molec Imaging 2012;7:26-34.

13. Li Y, Sheth VR, Liu G, Pagel MD. A self-calibrating PARACEST MRI contrast agent that detects esterase enzyme activity. Contrast Media Molec Imaging 2011;6(4):219-228.

14. Yoo B, Pagel MD. An overview of responsive MRI contrast agents for molecular imaging. Front Biosci 2008;13:1733-1752.

15. Hingorani DV, Bernstein AS, Pagel MD. A review of responsive MRI contrast agents: 2005-2014. Contrast Media Molec Imaging 2015;10(4):245-265.

16. Yoo B, Pagel MD. A PARACEST MRI contrast agent to detect enzyme activity. J Am Chem Soc 2006;128:14032-14033.

17. Liu G, Li Y, Pagel MD. Design and characterization of new irreversible responsive PARACEST MRI contrast agent that detects nitric oxide. Magn Reson Med 2007;58:1249-1256.

18. Sheth VR, Li Y, Chen LQ, Howison CM, Flask CA, Pagel MD. Measuring in vivo tumor pHe with CEST-FISP MRI. Magn Reson Med 2012;67:760–768.

19. Yoo B, Sheth VR, Howison CM, Douglas MJK, Pineda CT, Maine EA, Baker AF, Pagel MD. Detection of in vivo enzyme activity with catalyCEST MRI. Mag Reson Med 2014;71(3):1221-1230.

20. Liu G, Li Y, Sheth VR, Pagel MD. Imaging in vivo extracellular pH with a single PARACEST MRI contrast agent. Molec Imaging 2012;11:47-57.

21. Daryaei I, Pagel MD. Double agents and secret agents: the emerging fields of exogenous chemical exchange saturation transfer and T2-exchange magnetic resonance imaging contrast agents for molecular imaging. Res Rep Nuc Med 2015;15:19-32.

22. Soesbe TC, Ratnakar SJ, Milne M, Zhang S, Do QN, Kovacs Z, Sherry AD. Maximizing T2-exchange in Dy(3+)DOTA-(amide)X chelates: fine-tuning the water molecule exchange rate for enhanced T2 contrast in MRI. Magn Reson Med 2014;71:1179-1185.

23. Daryaei I, Randtke EA, Pagel MD. A biomarker-responsive T2ex MRI contrast agent. Magn Reson Med, 2017, Epub ahead of print. PMID: 27090199, DOI: 10.1002/mrm.26250

24. Soesbe TC, Merritt ME, Green KN, Rojas-Quijano FA, Sherry AD. T(2) exchange agents: a new class of paramagnetic MRI contrast agent that shortens water T(2) by chemical exchange rather than relaxation. Magn Reson Med 2011;66(6):1697-1703.

Proc. Intl. Soc. Mag. Reson. Med. 25 (2017)