Next Article in Journal
Alpha-Glucosidase- and Lipase-Inhibitory Phenalenones from a New Species of Pseudolophiostoma Originating from Thailand
Previous Article in Journal
Chemical Composition, Algicidal, Antimicrobial, and Antioxidant Activities of the Essential Oils of Taiwania flousiana Gaussen
Previous Article in Special Issue
Optimization of Cation Exchange for the Separation of Actinium-225 from Radioactive Thorium, Radium-223 and Other Metals
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Step-by-Step Guide for the Novel Radiometal Production for Medical Applications: Case Studies with 68Ga, 44Sc, 177Lu and 161Tb

by
Zeynep Talip
1,*,
Chiara Favaretto
1,2,
Susanne Geistlich
1,2 and
Nicholas P. van der Meulen
1,3
1
Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland
2
Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
3
Laboratory of Radiochemistry, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland
*
Author to whom correspondence should be addressed.
Molecules 2020, 25(4), 966; https://doi.org/10.3390/molecules25040966
Submission received: 30 January 2020 / Revised: 16 February 2020 / Accepted: 17 February 2020 / Published: 20 February 2020

Abstract

:
The production of novel radionuclides is the first step towards the development of new effective radiopharmaceuticals, and the quality thereof directly affects the preclinical and clinical phases. In this review, novel radiometal production for medical applications is briefly elucidated. The production status of the imaging nuclide 44Sc and the therapeutic β--emitter nuclide 161Tb are compared to their more established counterparts, 68Ga and 177Lu according to their targetry, irradiation process, radiochemistry, and quality control aspects. The detailed discussion of these significant issues will help towards the future introduction of these promising radionuclides into drug manufacture for clinical application under Good Manufacturing Practice (GMP).

1. Introduction

Cancer is regarded as one of the main health problems faced in the world today. Recent reports show that more than 10.000 hospitals worldwide use radionuclides for in vivo diagnosis or therapy and about 35 million patients receive cancer therapy using radionuclides annually [1]. Other than cancer, radionuclides are used in medicine for the diagnosis and treatment of various diseases, such as cardiovascular and brain disorders. It is expected that the demand for medically-applied radionuclides will dramatically increase in the years to come. According to the recent Fior Markets report, the global radiopharmaceuticals/nuclear medicine market is expected to grow from USD 6.5 billion in 2017 to USD 12.41 billion by 2025 [2].
Different radionuclides are applied in various fields of nuclear medicine. Taking the radionuclide in question’s half-life and decay emission into consideration, they are used for imaging, via positron emission tomography (PET) or single-photon emission computed tomography (SPECT), and for therapy via α-, β-, or conversion and/or Auger electron emission. Beta particles, alpha particles, and Auger electrons can irradiate tissue volumes with multicellular, cellular and subcellular dimensions, respectively [3,4] thanks to the different linear energy transfer (LET), defined as the amount of energy transferred by a traveling particle to the surrounding material per unit of length. In particular, it is of great interest to develop clinical methods using conversion/Auger-electron-emitting radionuclides towards the treatment of disseminated tumor cells and small metastases. To date, clinical research in this regard has been very limited and there is still much to learn about the Auger electrons as a potential therapeutic [3,4,5,6,7].
Radiometals have been used extensively in clinical diagnostics over the last three decades, particularly, 99mTc, 67Ga and 111In for SPECT and, more recently, 68Ga, 89Zr and 64Cu for PET (Table 1). On the other hand,, therapeutic radiometals in clinical use are limited to 177Lu [8,9,10], 90Y [11,12], 89Sr [13,14] and 223Ra [15,16]. 223Ra is the only approved radionuclide by health authorities for targeted alpha therapy to extend survival [17]. It is worth mentioning that to bring a novel radionuclide from the development stage to the clinic is a long process. Phase III data for 177Lu demonstrated that radioactive drugs took more than 15 years to achieve the European Medicines Agency (EMA) and US Food and Drug Administration (FDA) approval [18]. So far, only 177Lu-DOTATATE has been approved for clinical use, while four other 177Lu-based radiopharmaceuticals have been clinically tested [19,20,21].
In the last decade, terbium and scandium radionuclides have received special attention for their potential use in radiopharmacy. The therapeutic β--emitter 161Tb and the diagnostic radionuclide 44Sc are considered as an alternative to 177Lu and 68Ga, respectively. 161Tb can be prepared in high specific activity and it has lower energy γ-emission (48.9 keV (17%), 74.6 keV (10.2%)) than its 177Lu counterpart. Dose calculations have shown that 161Tb can be more effective for very small tumor lesions due to the emission of low-energy conversion and Auger electrons [23,24]. Moreover, preclinical in-vivo and in-vitro comparison studies with DOTA-folate conjugate and prostate-specific membrane antigen (PSMA)-617 labeled 177Lu and 161Tb have shown better results for 161Tb [25,26,27]. Marin has reported that the SPECT images of 161Tb are comparable to those of 177Lu using particular settings of the SPECT device in question [28]. Another attractive property for terbium is that, other than 161Tb, it has two diagnostic radioisotopes 152Tb (T1/2 = 17.5 h, Eβ+av: 1140 keV) for PET and 155Tb (T1/2 = 5.32 d, Eγ: 87 keV (32%), 105 keV (25%)) for SPECT, making it ideal for the “matched pair” principle of theranostics [29,30].
44Sc has promising decay characteristics (T1/2 = 4.04 h, 94.27% β+ and 5.73% EC, Eβ+av: 632 keV), which makes it a good alternative to the short-lived 68Ga (T½ = 67.71 min, 89% β+ and 11% EC, Eβ+av: 830 keV). Since the chemistry of 44Sc is similar to those of the lanthanides, it is proposed to be used as a diagnostic match to 177Lu and for pre-therapeutic dosimetry studies. Moreover, 44Sc has a theranostic β- emitting matched pair 47Sc (T1/2 = 3.35 d), which is particularly interesting for radionuclide therapy [31,32,33]. Furthermore, the differences between the coordination chemistry of 68Ga and 177Lu cause deviation in pharmacokinetic studies [34,35]. 44Sc has better spatial resolution for PET imaging than 68Ga, thanks to its lower positron energy [36]. In addition, its longer half-life (T1/2 = 4.04 h) [37] allows performing late-time-point PET imaging and its transfer to PET centers, while 68Ga only allows image-taking over a few hours.
This review is divided into two parts: part one briefly covers the required steps for the production of novel radionuclides towards medical application, while part two outlines the status of the production of two emerging novel radionuclides, namely, 44Sc and 161Tb. Their progress is explained in detail in comparison with the clinically-applied radionuclides, 68Ga and 177Lu, respectively.

2. Radionuclide Production: A Step-by-Step Guide

The production of novel radionuclides for innovative radiopharmaceuticals requires multidisciplinary collaboration between nuclear physicists, material scientists, radiochemists, engineers, radiopharmacists, immunologists, structural biologists, health physicists, coordination chemists, etc. Since radionuclide production is the first step of this chain, it requires an interface between all these disciplines for the radionuclide in question to reach the clinic.
The production of novel radionuclides for medical applications essentially consists of four successive steps: targetry and irradiation of targets (including cross-section measurements and production rates), radiochemistry (chemical separation) and quality control (Figure 1). Production of sufficient amounts and high quality of novel radionuclides requires systematic research in each of these steps. The selection of radionuclide candidates requires careful considerations based on several factors:
1)
Radiation characteristics such as the type(s) of decay emission(s), the energy of the emission(s), LET, physical half-life. If a gamma-ray photon is emitted in the decay process, its energy and branching ratio should be in the diagnostically useful range (dose received by the patients should be minimized, photon energy should be mostly less than 400 keV [38]). The physical half-life of the therapeutic radionuclides is extremely important for minimizing decay loss and provides logistical advantages for the shipment. Most of the novel radionuclides have relatively long half-lives, however, in some cases, due to their short half-life, irradiation facilities should be on-site.
2)
The cost of the target material and stable long-term supply.
3)
Availability of the facility (production capability and logistics).
4)
A reasonable production cost.
5)
The chemical separation method should be as simple as possible, for remote handling and to produce no-carrier-added (n.c.a.) products.

2.1. Targetry

Targetry is the first step for radionuclide production. Currently, the sources of the radionuclides produced are nuclear research reactors and cyclotron facilities using protons, deuterons and alpha particles. The use of electron linear accelerators are on the rise. Radionuclide generators are also commonly used; however, they still need a reactor or cyclotron source to produce the parent radionuclide. The choice of target material for the production is key, as the irradiation parameters, as well as the subsequent devised chemical separation, are affected.

2.1.1. Target Preparation for Cyclotrons

Solid Targets

Most novel radiometals are produced using solid target systems. Methods for solid target manufacture are usually classified as mechanical, physical and chemical. Several different methods such as sintering, rolling, melting, sedimentation, mechanical pressing, electrodeposition, magnetron sputtering, etc. are used for the cyclotron solid target preparation [39,40]. The use of mechanical methods (rolling and pressing) is most common, which consists of a mechanical reshaping of solid or powder materials to produce self-supporting targets. The following criteria are important for the design of the solid target to be irradiated:
1) Selection of the target and backing material: target and backing material are chosen based on high stability and high melting point. In addition, target material should have a good heat conduction capability at the interface of the target and backing material to have efficient heat transport. Targets must be mechanically stable and resistant to radiation damage. The chemical form of the target material should be compatible with the post-irradiation processing, while backing material (if used) should be chemically inert in the respective dissolution process. Target material must be as chemically and isotopically pure as reasonably possible. Impurities can enhance the production of undesired side products, which could impact the radionuclidic purity and subsequent labeling process. In addition, target uniformity, homogeneity and contaminants strongly depend on the target preparation method.
2) Target thickness: target thickness is another important parameter for radionuclide production. It is possible to minimize the side products, depending on the chosen energy range and the thickness of the target. Optimal target thickness can be evaluated using SRIM software by taking the irradiation angle of the beam into consideration [41].
3) Efficient cooling: when targets are irradiated, it generates heat. Cooling systems should be such that the irradiated target retains its integrity as much as possible. Especially for target materials with a low melting point, the surface temperature of the target can be critical.

Liquid Targets

Liquid target systems, while popular for the production of 18F using enriched water, are also used for the production of radiometals such as 68Ga, 64Cu, 61Cu, 89Zr, 44Sc, 86Y,63Zn and 94mTc [42,43]. They can shorten the pre- and post-irradiation target preparation steps and simplify the target transfer after irradiation [42]. In addition, liquid targets have the advantage of producing radiometals with existing medical cyclotrons without investment for a solid-target station [44]. Liquid targets should be prepared such that saturation point isn’t reached such that precipitation occurs.

2.1.2. Target Preparation for Reactors

When using reactors to irradiate target material, they are encapsulated in high purity quartz ampoules in powder form. Before irradiation, the quartz ampoule should be carefully cleaned to avoid the activation of potential impurities. The target material is often initially dissolved, placed into the ampoule and carefully evaporated to dryness. The ampoule is sealed and placed in an aluminum capsule, which is also sealed by electron-beam welding. The chemical purity of the product strongly depends on the opening of the quartz ampoule [45]. The low cross-section data of a radionuclide imposes the use of as large amounts of target material as reasonably possible. Small amounts of target material can also be compensated by high neutron beam flux of the reactor, however.

2.2. Production

Production yield depends on the beam flux, energy, irradiation time and the number of target nuclei, respectively. To determine the most efficient production routes and to minimize the number and amount of side products, nuclear reaction cross-section data of the target material have crucial importance. It should be pointed out that the inherent success of any production strategy requires precise nuclear data [46,47,48]. Between 2012 and 2016, extensive nuclear data studies were carried within a research project, coordinated by the International Atomic Energy Agency (IAEA) consisting of compilation, evaluation, and recommendation of cross-section data for the production of medical radionuclides [49,50]. As a result, considerable extensions and valuable improvements have been made to the IAEA-NDS recommended cross-section database for the production of PET and gamma-emitting radionuclides.
It is important to investigate all the possible production routes and then to choose the most proper reaction and energy range. However, even after optimization of all the parameters, the experimental production yields are lower than the theoretical thick target yields due to several factors such as loss of beam particles, radiolysis, variation of the beam intensity, density reduction and/or evaporation in the beam path, etc. [51,52].

2.2.1. Cyclotrons

Neutron deficient radionuclides are produced using a cyclotron and they decay by electron capture (EC) or β+-emission. The majority of radionuclides produced using cyclotrons utilize the “Target Material (p,xn)Desired Radionuclide” nuclear reaction. Cyclotrons are classified into three categories, namely, small medical cyclotrons (< 20 MeV), intermediate-energy cyclotrons (20–35 MeV) and high-energy cyclotrons (>35 MeV). According to the report of Goethals and Zimmermann 1050 small medical cyclotrons, 100 intermediate-energy cyclotrons and 50 high-energy cyclotrons were reported worldwide in 2015 [53].
Depending on the type of cyclotron being used, the proton energy can be degraded to lower energies with selected materials to be able to investigate different production routes [54]. In some facilities, deuterons and helium also have been used as a projectile. However, their availabilities are limited compared to proton accelerators.

2.2.2. Nuclear Reactors

Neutron-rich radionuclides are produced by nuclear reactors and they decay by βemission. The majority of radionuclides produced using nuclear reactors utilize the “Target Material (n,γ)Desired Radionuclide” nuclear reaction. Nuclear research reactors are also classified into three categories, namely, low neutron flux (<1013 cm−2s−1), medium neutron flux (2–8 × 1013 cm−2s−1) and high neutron flux (>1014 cm−2s−1) reactors. Several imaging or therapeutic medical radionuclides (99Mo/99mTc, 90Y, 131I, 153Sm, 166Ho, 177Lu, 161Tb, etc.) are currently produced using nuclear reactors.
According to the International Atomic Energy Agency (IAEA) Research Reactor Database, 97 research reactors are reported to be in use towards radionuclide production [55]. Krijger et al. reviewed the status and necessity of nuclear reactors for targeted radionuclide therapy [56]. The availability of the high thermal neutron flux (>1014 cm−2s−1) nuclear research reactors is especially important for the production of medical radionuclides with high specific activities. Most of the nuclear research reactors currently in service for this purpose, however, were built in the 1960s and they require longer and more frequent shut-down periods for maintenance to continue operation with minimum failure, which limits their radionuclide production capability considerably. Considering the expanding growth of the nuclear medicine market, it is foreseen that the capacity of the remaining nuclear research reactors will not fulfill the demand for therapeutic radionuclides in the near future.

2.2.3. Alternative Production Routes

In some cases, it is difficult to produce carrier-free or radionuclidically pure products with conventional reactor-based or accelerator-driven production routes. In this case, the use of mass separators to produce carrier-free radionuclides for nuclear medicine is becoming more and more attractive. However, this method requires large-scale mass separation facilities which are available for this purpose in few places worldwide: MEDICIS at CERN [57,58,59,60,61], ISAC at TRIUMF, Canada [62], FRIB (under construction in Michigan, U.S.A.), ISOL at MYRRHA (under construction in Mol, Belgium) [63] and SPES-INFN (under construction in Legnaro, Italy). At these facilities, radionuclides are produced via high-energy proton induced reactions combined with an online mass separator, known as Isotope Separation On-Line (ISOL). Alternatively, it has been considered to use external proton or neutron-irradiated targets to perform offline mass separation to enhance the possibilities of producing promising novel radionuclides with the mass separation technique [64]. The mass separation technique is based on the ionization processes such as surface, plasma or laser ionization, however, these methods are not element selective and could cause isobaric contamination in the resultant material. As a result, radiochemical separation processes must still be developed to use mass-separated radionuclides for nuclear medicine applications. Other than cyclotron and nuclear reactor production routes, there are also much less common alternative production routes such as the use of electron linear accelerators [65,66], Van der Graaf accelerators [67] or lasers [68,69,70] to produce radionuclides for nuclear medicine applications.

2.3. Radiochemistry

Radiochemistry is used for two purposes, to isolate the desired radionuclide in n.c.a. form, by means of chemical separation, and to recover the enriched target material for reuse.
Different chemical separation techniques have been considered to obtain n.c.a. radionuclides, such as ion-exchange chromatography, extraction chromatography, solvent extraction, precipitation, and electrochemical methods. The selection of the chemical separation process depends on several parameters:
1)
Adaptation of the separation using a remote/semi-automated operation system;
2)
Separation processing time;
3)
Product yield as a result of the separation process;
4)
Recovery of enriched target material for recycling;
5)
The chemical form of the final product;
6)
Acidity and the volume of the final product;
7)
Robustness and reproducibility of the process.
Ion exchange and extraction chromatography are the most convenient methods for the remote/semi-automated separation and purification operations. In this case, distribution of an element between the solution and ion exchange resin, which is defined as distribution coefficient data, has a great value to define the separation process (Figure 1).
The processing of the irradiated target material requires hot cells in order to decrease the radiation exposure of the operators. Removal of the irradiated target from the target station is performed remotely and is transferred to a reception hot cell. It is very important to eliminate the risk of contamination during target removal and in the processing thereof [71].

2.4. Quality Control of the Radionuclide for Preclinical Studies

After radiochemical separation, the quality of the final product requires assessment. This is a vital step for the development of novel radionuclides towards nuclear medicine applications. As part of the quality control process, radiochemical purity, specific activity, radionuclidic purity, and chemical purity measurements should be performed and radiolabeling processes should be established.
Radiochemical purity is defined as the fraction of the total radioactivity of the radionuclide concerned in the sample, which is present as its desired chemical form. It is generally tested by radio-chromatographic methods, such as thin-layer chromatography (TLC) or high-performance liquid chromatography (HPLC).
Specific activity is defined as radioactivity per unit mass of the product [72]. Several factors that could affect the specific activity of the final product include cross-sections, target impurities, secondary nuclear reactions, target burnup, and post-irradiation processing periods [20].
Accurate activity measurement is crucial for activity dosing and absolute image quantification. According to the international guidelines, the dose of radiopharmaceuticals should be measured with a minimum accuracy of 5 to 10% [73,74,75]. Radionuclide calibrators consisting of ionization chambers are used to measure the product activity. They can be used over a wide range of activities from hundreds of kBq up to tens of GBq. Typically, calibration factors are determined by direct measurement of the samples with a primary standard. Moreover, precise nuclear data information such as decay emission(s), the energy of the emission(s) and half-life are important to perform precise activity measurement.
Activity measurement also requires regular quality control tests (accuracy, geometry, linearity, and constancy) to avoid wrong activity measurements. Geometry effects (same reading for the same amount of activity irrespective of the volume or the vial of the sample) can dramatically affect the activity measurements, particularly for low-energy X-ray or gamma photons. It should be, therefore, assessed for the vials used to collect the product, as well as those used for quality control and in GMP [76,77].
Radionuclidic purity is defined as the ratio of the radioactivity of the desired radionuclide to the total radioactivity content of the sample. Gamma spectroscopy is the main instrument used for the determination of the radionuclidic purity. Efficiency calibration of the detector system must be properly performed with calibrated standard gamma sources. Measurements should be repeated after an appropriate decay time to determine long-lived contaminants.
Chemical purity is controlled by the determination of the impurities in the final product. It has explicit importance and has a direct effect on the radiolabeling of the final product to a target molecule. The macrocyclic chelators are not selective for the radiometal of interest and they can coordinate with other (nonradioactive or “cold”) metal ions and depends on the specific activity of the final product, where even ppb level impurities could affect the labeling efficiency [78,79,80]. The shelf life of the product also strongly depends on the chemical purity and specific activity. Other than the initial chemical impurities, the effect of decay products on labeling efficiency should also be investigated. Different analytical techniques such as ICP-OES, ICP-MS, ICP-AES etc. could be used to determine the chemical purity of the final product.
Radiolabeling efficiency is defined as the ability of a chelator to form its radionuclide complex. The final product is mixed with a chelator-(peptide/antibody) and pH is adjusted with a suitable buffer solution. The resultant solution is then heated for a certain time and temperature, based on the radiolabeling kinetics of the chelator used. Several reaction parameters such as the concentration of peptide or antibody, buffer conditions, pH, reaction temperature and reaction time can affect the labeling efficiency. In particular, pH plays a critical role in radiolabeling, particularly on the rate of complex formation. In addition, radical scavengers such as gentisic acid and ascorbic acid are used to increase the stability of the labeled compound. Ethanol has also been used to improve the radiolabeling efficacy and to prevent radiolysis [81].

2.5. Automation of the Labeling Procedure Towards GMP Production

Radiopharmaceutical products for clinical applications must follow the universal requirements for the drug products. Drug products must for the sake of patient safety be of good quality and have a favorable risk-benefit balance when overall efficacy and safety are assessed. The European Pharmacopoeia (EP) and likewise the United States Pharmacopoeia (USP) and the Japanese Pharmacopoeia (JP) set the minimum quality standards for drug products in general and also for radiopharmaceuticals. When developing new radionuclides intended to be used in new drug products, these monographs should be considered amongst others: dosage form - EP 0520 “Parenteral Preparations”; drug product classes - EP 0125 “Radiopharmaceutical Preparations” and for specific guidance in quality control of such products EP 2.2.66. “Detection and Measurement of Radioactivity”. Recently the European Directorate for the Quality of Medicines & HealthCare (EDQM) published the “Guide for the elaboration of monographs on Radiopharmaceutical Preparations” [82], which is a useful resource for setting quality parameters and specifications for new radiopharmaceuticals.
Apart from the pharmaceutical regulations, it is of key importance to also consult the national radioprotection legislation. The endeavor to translate new radionuclides into the clinic is certainly a procedure that involves many steps. When planning for the first-in-man clinical trial, the quality of labeling solution, as well as overall product quality, should be assessed against EP quality standards to guarantee a good quality product that reproducibly meets all the predefined specifications. When evolving towards later stages of clinical studies and especially towards commercialization of a drug product, full GMP will apply as published by the European Commission’s EudraLex volume 4 of “The rules governing medicinal products in the European Union” [83].
Once a novel radionuclides such as 161Tb or 44Sc is introduced into a first clinical trial all changes in the manufacture of radionuclide and drug products with potential impact on the quality of the product should go through a formal internal assessment before such changes are applied. Such changes may be an improvement of the targetry, change of the supplier of the target material, changes of solvents or their quality or change of the drug product vial. The assessment should cover aspects like re-validation of processes and quality control (QC) methods, adaptation of specifications and overall impact on the quality of the drug product. Product specifications include [83]:
1)
appearance;
2)
pH;
3)
radiochemical purity/yield;
4)
radionuclide identity/radionuclidic purity;
5)
radioactivity concentration;
6)
tracer identity/quantification;
7)
molar activity;
8)
chemical purity;
9)
residual solvents;
10)
bacterial endotoxins (BET);
11)
sterility;
12)
product stability;
To verify compliance with the specifications, controls must be conducted by suitable and documented procedures for starting materials, including the radionuclide precursors, and final products. Moreover, controls have to be operated also on equipment that has been qualified for the tests to be applied to the actual radiopharmaceutical or radionuclide produced [83]. The validation characteristics such as accuracy, precision, specificity, detection limit, quantification limit, linearity, and range should be considered [82]. QC is that part of GMP which is concerned with sampling, the mentioned specifications of each quality parameter (see above) and with the organization, documentation, and release procedures which ensure that the necessary and relevant tests have been actually and successfully carried out [84]. All the test methods require validation and records to demonstrate that all the steps (sampling, inspecting, testing procedures) were properly carried out and assessed against the defined specifications. Pharmaceuticals are not released to the clinics for the patient use until their quality has been shown to fulfill the required quality. However, in the case of radiopharmaceuticals QC with ultra-short-lived radioisotopes (i.e., positron-emitting radioisotopes 11C, T1/2 = 20 min; 18F, T1/2 = 110 min, etc.), it is necessary to administer the product before all the tests have been completed. As described earlier, a very important requirement is that records of all the controls should be reported [82].
Automated modules are regularly used to ensure consistency of labeling the radionuclide to the target to produce the radiopharmaceutical. The so-called GMP module is utilized for the pharmaceutical labeling of a limited number of radionuclides for clinical diagnosis and therapy, namely, 18F, 11C, 15O, 13N, 68Ga, 99mTc, 90Y and 177Lu [85,86,87,88].
Stringent regulatory demands make fully-automated approaches necessary for the synthesis of radiopharmaceuticals to be used in clinics. In addition, an automated system can minimize human intervention and guarantee better control of sterility and pyrogenicity of radiopharmaceuticals [89]. A current trend in the development of automated systems is moving from the fixed tubing system, that have characterized a large variety of synthesis modules over the years, to a “disposable sterile cassette”-based system [90]. The disposable cassette systems are intended for single-use and are designed for one or more specific syntheses. 68Ga/177Lu/90Y/111In DOTA-peptides have been synthesized with high purity, high radiochemical yield and acceptable synthesis time using such a system [91].
The development of an automated procedure for therapeutic radiolabeling with novel radionuclides is a considerable challenge and several factors have to be taken into consideration. Firstly, the final product has to be purified from possible free activity (radionuclide that is not associated with the conjugate molecule), which is performed using cartridge-based methods. Furthermore, to achieve high labeling efficiencies of conjugates, the concentrations of the reagents used must be high and, therefore, low volumes are used. These small volumes of reagents require transfer through the system without or minimal loss of solution. Another factor to keep in mind is the number of reagents that a radiolabeling procedure typically requires [92]:
1)
the conjugate molecule;
2)
the radionuclide;
3)
a buffer to maintain pH;
4)
a scavenging agent (such as ascorbic acid) to prevent radiolysis;
5)
a quenching agent (such as EDTA) to complex any radionuclide that is not associated with the conjugate molecule;
6)
diluent to formulate the preparation for administration to patients.
It is also important to prevent contamination of the product with metallic impurities, which may compete with the radionuclide for the limited number of chelating sites on the conjugate molecule [79]. This implies that metallic transfer systems (such as needles) ought to be avoided where transferring the acidic radionuclide solutions (pH 1–2). In parallel to the development of the automated synthesis procedure, the appropriate quality controls have to be established and validated. In the following section, the current status and the recent developments for the production of 44Sc and 161Tb will be presented in comparison with 68Ga and 177Lu, respectively.

3. The Diagnostic Radionuclides 68Ga and 44Sc

3.1. Targetry/Production

Table 2 shows the comparison of 68Ga and 44Sc and decay characteristics and production routes. To date, several different target materials are considered for 68Ga, 44Sc production (Table 3).

3.1.1. 68. Ga Generator Production

Commercially available 68Ge/68Ga generators are produced via 69Ga(p,2n)68Ge reaction by irradiating natGa with 23 MeV proton energy [89]. The 68Ge (T1/2 = 270.95 d) produced is separated from the Ga target material [105,106] and the final product loaded onto an inorganic substrate (SnO2 or TiO2) which is shielded with Pb. 68Ge decays by electron capture to 68Ga (T1/2 = 67.71 min) and the daughter is subsequently eluted from the resin using dilute HCl (0.1 -0.6 M HCl) which is introduced to a GMP module for radiolabeling. Alternatively, 68Ge can be produced with high energy protons utilizing the 71Ga(p, 4n)68Ge reaction or with an alpha beam via the 66Zn(α, 2n)68Ge nuclear reaction [107].

3.1.2. 68. Ga Cyclotron Production

Solid targets: production of 68Ga is also possible by using enriched 68Zn or natZn targets using the 68Zn(p,n)68Ga nuclear reaction with a medical cyclotron at ~12 MeV (to ensure no 67Ga production via the (p,2n) reaction at >12 MeV) [101]. However, even using enriched 68Zn targets, the co-production of 66Ga (T1/2: 9.49 h) and 67Ga (T1/2: 3.26 d) cannot be avoided. Previously, electroplating of 68Zn on different backing materials such as copper, silver and platinum were reported [101,102,103]. Using natural copper and silver as a backing material would lead to the production of long-lived 65Zn (T1/2 = 244 d) and 109Cd (T1/2 = 453 d) via the 65Cu(p,n)65Zn and 109Ag(p,n)109Cd nuclear reactions, respectively. Platinum backing has a lower thermal conductivity than copper and silver, however, it is considerably more expensive. 68Zn foil targets can also be used directly for 68Ga production [98], nevertheless, commercial availability of enriched Zn foil is limited. In a recent study, enriched 68Zn pressed targets were also used for the large-scale production (up to 140 GBq) of 68Ga [108]. Effective molar activity was reported as 77.4 ±5.0 GBq/μmol with 89% recovery yield.
Liquid targets: alternatively, the production of 68Ga by using liquid zinc chloride and zinc nitrate targets with medical cyclotrons at ~ 13-14 MeV have been reported [42,93,95,96,97]. Zinc chloride solution can produce high target pressure during irradiation by increasing gas evolution and it is highly corrosive [97]. On the other hand, nitrate ions decrease the gas evolution by increasing the hydroxyl radical formation in the solution, which makes it a better option compared to zinc chloride targets. Alveset al. reported that up to 24 TBq/µg specific activities of 68Ga can be reached by using liquid 68Zn(NO3)2 targets [42].
Cyclotron production of 68Ga has the advantage of extremely high activities of 68Ga over eluting product from a generator.44. Sc generator production

3.1.3. 44Sc Generator Production

44Sc generators are produced via the 45Sc(p,2n)44Ti nuclear reaction.44Ti (T1/2 = 60.6 a) decays by electron capture to 44Sc, which subsequently decays to stable 44Ca. The advantage of this method is the target material 45Sc, which is the only naturally occurring stable isotope of scandium; therefore, it does not require expensive enriched target material for irradiations. This reaction has a low production rate and requires high proton flux and a long irradiation time for the production of 44Ti with high radioactivity yields [109,110]. Production of 185 MBq 44Ti was reported with an internal proton beam of 28.5 MeV and 60 and 500 µA external and internal proton fluxes [111].
Alternatively, 44Ti can be produced with high energy protons using vanadium or iron targets thanks to spallation reactions (51V(p,2p6n)44Ti and 54Fe(p,11B)44Ti). This process requires a complex radiochemistry for the purification of 44Ti from proton irradiated target material, however [112,113,114].
A 370 MBq (10 mCi) generator should be able to be eluted every 4 h to provide 148-185 MBq (4–5 mCi)—the equivalent of a single patient dose [115]. In addition, the lifetime of 44Ti/44Sc generator is longer than the one of 68Ge/68Ga due to the long half-life of 44Ti. Nevertheless, the co-production of longer-lived radionuclidic impurities such as 48Sc (T1/2 = 43.7 h) and 44mSc (T1/2 = 58.5 h) would cause an additional dose burden to the patients.
Generator-produced 44Sc has been used for preclinical investigations [116] and the first 44Ti/44Sc generator used for such purpose achieves elution of 180 MBq 44Sc with 97% yield and the breakthrough of 44Ti was reported as 90 Bq. [109]. In 2012 the first clinical application of [44Sc]Sc-PSMA-617, utilized with generator-derived 44Sc, occurred for PET imaging of metastasized castrate-resistant prostate cancer, as a proof-of-principle study [35]. The qualities of the images obtained with [44Sc]Sc-PSMA-617 were comparable with those of [68Ga]Ga-PSMA-11.

3.1.4. 44. Sc Cyclotron Production

Solid targets:44Sc production via proton irradiation of natural metallic calcium targets (natCa(p,n)44Sc) was investigated by Severin et al. and Valdovinos et al. [117,118]. Severin et al. reported a 44Sc production yield of more than 650 MBq with a 70% yield. This process does not need target recycling, however, radionuclidic purity of the final product (95%) is not regarded as sufficient for clinical application. Other than the relatively low production yield, the use of natural calcium leads to the co-production of long-lived radionuclides, namely, 44mSc, 47Sc (T1/2 = 80.4 h) and 48Sc, which can lead to additional dose burden to patients.
The use of enriched 44Ca targets was considered a better option for 44Sc production (44Ca(p,n)44Sc), available commercially in carbonate form [99,100]. One of the drawbacks of using CaCO3 as a target material is the release of CO2 during irradiation, which could cause the distortion of the target capsule. To avoid this from occurring, enriched 44CaCO3 was annealed at 900oC to convert it to 44CaO, which was then used as the target material [104].
Alternatively, Duchemin et al. and Alliot et al. investigated the production cross-section of deuteron-irradiated 44Ca targets (CaCO3) [119,120]. Alliot et al. demonstrated that 44Sc production with deuterons leads to higher cross-section and higher radionuclidic purity in comparison to that of proton irradiation [120]. Production of Sc radionuclides via α-particle irradiation was also investigated and it was demonstrated that the ratio of 44mSc/44Sc is five and twenty times higher in comparison with deuteron and proton-induced reactions, respectively [121].
Liquid targets: Production of 44Sc with liquid targets (natural Ca(NO3)2 in ultrapure water) was investigated utilizing the 44Ca(p,n)44Sc nuclear reaction. [94]. The production yields reported (28 ± 1 MBq) were much lower than those reported using solid targets.

3.2. Radiochemistry

3.2.1. 68. Ge/68Ga Generator Production

The success of radionuclide generators depends on the selection of the parent/daughter pair and the radiochemical separation procedures. Dash et al. summarized the selection criteria for the radiochemical separation method for radionuclide generators [122]. Radiochemical separation method should be fast, reproducible and provide the daughter radionuclide in high radionuclidic, radiochemical and chemical purity.
The 68Ge product is subsequently loaded onto a shielded column containing, predominantly, SnO2 or TiO2 as matrix – to create what is known as a generator system. The 68Ge is left on the column matrix to decay to its daughter, 68Ga, which is then eluted (or “milked”) as 68Ga3+ with 0.1 M HCl. The breakthough of 68Ge from the TiO2 column is reported as ~ 10−3 % of the eluted 68Ga activity [123]. Due to the presence of metallic impurities such as Fe(II), Mn(II) and Zn(II), post processing of the eluate (68GaCl3) is required for high labeling yield and high specific activity [124].

3.2.2. 68. Ga Cyclotron Production

Target material should be processed to remove the bulk zinc and other metal impurities prior to radiolabeling process of 68Ga. To date, several different separation techniques such as ion exchange chromatography, solvent extraction [125,126], thermal diffusion [127] and precipitation [128] techniques are used for the separation of Zn from Ga. Two-step column separation systems have been considered for the ion exchange method. The first column (AG50W-X8 cation exchange resin or Hydroxamate) has been used to separate macro quantities of Zn from 68Ga, while the second column (AG1-X8 anion exchange resin, DGA or UTEVA extraction resins) is used to further reduce any traces of metallic impurities, while also decreasing the acidity and the volume of the final product [96,97,103]. Comparison of the chemical purities in generator- and cyclotron produced 68Ga solutions showed that zinc content was significantly lower for cyclotron-produced 68Ga [103].

Recycling of 68Zn

The price of commercially available enriched 68Zn is relatively low. As a result, the facilities utilizing this method do not currently recycle the target material [103].

3.2.3. 44. Sc Generator Production

Due to the long physical half-life of 44Ti, the long-term stability of the generator is essential. The separation process should provide high 44Sc elution and zero-to-low 44Ti breakthrough. The radiochemistry for the generator system consists of a column containing AG1-X8 anion exchange resin. 44Ti is dissolved in 0.1 M H2C2O4 and loaded onto the column. Once secular equilibrium was reached 44Sc could be eluted with 20 mL of 0.005 M H2C2O4/0.07 M HCl solution, with >97% elution yield (180 MBq) and low 44Ti breakthrough (5 × 10−5 % or 90 Bq) [109]. It was discovered, however, that the eluate was too dilute and acidic for direct labeling. As a result, a post elution processing step was developed, with purified 44Sc eluted with 3 mL 0.25 M ammonium acetate buffer (pH = 4.0) [110].
A gas-phase separation method for the separation of scandium from titanium foil has also been proposed, via thermal release in vacuum [129]. It was shown that by heating around 1200 °C for one hour, almost all the scandium is released from titanium matrix.

3.2.4. 44. Sc Cyclotron Production

Cyclotron production of 44Sc requires an efficient 44Sc/Ca separation procedure. Severin et al. reported separation of Sc by means of precipitation. After irradiation, natural calcium was dissolved in HCl and the Sc separated from Ca by its precipitation as Sc(OH)3 at neutral pH [117]. Different ion exchangers such as UTEVA, Chelex 100 and DGA resins are also commonly used to separate Sc from Ca [99,100,118,120], however, to date, DGA resin is the most commonly-used system for the separation of Sc [99,120]. It ensures elution of Ca in more concentrated HCl, while Sc can be eluted further with 0.1 M HCl. Van der Meulen et al. reported a fast and efficient Sc/Ca separation method with 98% efficiency [99]. After dissolving CaCO3 in 3.0 M HCl, 44Sc is separated from the target material using DGA resin, after which it is concentrated using SCX resin. Production of up to 2 GBq 44Sc was reported with high radionuclidic purity.

Recycling of 44Ca

The natural abundance of 44Ca (2.086%) is much lower than 68Zn (19.024%), therefore, recycling of the enriched 44Ca is important to decrease the cost of production. A simple recycling procedure was reported: firstly, calcium is precipitated as Ca-oxalate, after which it is converted to carbonate by slowly heating to 500 ºC [99].

3.3. Labeling Efficiency

3.3.1. 68. Ga

The end product for both cyclotron-produced and generator-eluted 68Ga is in chloride form. The main issue in the production process of 68Ga radiopharmaceuticals is the elution from the 68Ge/68Ga generator. While this is a popular means of obtaining the PET radiometal, there are some drawbacks using the 68Ga eluate for radiolabeling. The most pressing issues to be addressed are the large volume of the generator eluate (approximately 5 – 10 mL), the HCl concentration (0.1 – 1.0 M in metal oxide-based generators) and the presence of measurable activity of the long-lived parent radionuclide 68Ge (i.e., 68Ge breakthrough). Other metallic impurities generated from the generator columns (Ti4+), from the decay of 68Ga (Zn2+), or introduced as a contaminant in the process (Fe3+) may adversely affect the 68Ga labeling reaction, as well as the molar activity of the labeled product. As a result, the post-processing of the product is necessary, as mentioned previously. The eluate is loaded onto a cation exchange resin and the desired product separated from the impurities above [130]. The use of automated systems ensures high yields and safe preparation of 68Ga-labeled radiopharmaceuticals for routine application [90].
Schultz et al., developed a method for radiolabeling of DOTA-like conjugated peptides with generator-produced 68Ga, using NaCl-based cationic elution method. This method has an advantage of reducing preparation time, thanks to the absence of organic solvent in the eluent [87]. Although DOTA-peptides are the most widely used ones, it was shown that TRAP chelators have a better binding ability for 68Ga compared to DOTA and NOTA-peptides [131]. Similarly, under milder conditions, the synthesis of 68Ga(THP-TATE) is significantly faster compared to the 68Ga(DOTATATE) [132]. In a recent study, Sinnes et al., have shown that 68Ga(DATA-TOC) can be prepared under milder conditions than the 68Ga(DOTA-TOC), which has a practical advantage for instant kit-type labeling without effecting the efficacy [133].

3.3.2. 44. Sc

The optimum conditions for the labeling of 44Sc with DOTATOC, DOTA, and DOTATATE were reported in refs [134,135]. It was also demonstrated that 44Sc can be used for the labeling of biomolecules with both DOTA and NODAGA chelators [136]. It was reported elsewhere that DOTA is the most suitable ligand among a series of macrocyclic ligands for binding scandium radionuclides [137]. The stability study in the presence of rat serum demonstrated that 44m/44Sc-DOTATATE is stable over a period of up to 25 h [135]. Similar results were obtained by van der Meulen et al. for 44Sc-DOTANOC, which showed stability over 24 h. Moreover, an improved scavenging effect was observed with gentisic acid compared to ascorbic acid [99].
pH plays a critical role in radiolabeling of Sc, particularly on the rate of complex formation. Scandium is known to be a lanthanide-like element due to their similar chemistry, however, it forms different hydroxide complexes at the same pH in comparison to lanthanides. At pH 4.5 lanthanides are present only as Ln3+ [138], while Sc is present in different chemical speciations such as Sc3+, Sc(OH)2+, Sc(OH)2+ and insoluble Sc(OH)3 [139,140]. Taking this into consideration, optimal pH for Sc labeling should be lower than 4.5.

3.4. Production for the Clinical Applications

3.4.1. 68. Ga

The first pharmaceutical-grade 68Ga generator (Galli Ad) received a Marketing Authorization as a drug product by the EMA in 2014 [141]. Details of the commercially available 68Ge/68Ga generators are reported in [141]. The existing [68Ga]GaCl3 European Pharmacopeia monograph is based on the commercially available 68Ge/68Ga generators. Nowadays, many 68Ga-labeled tracers are used in clinical trials and NETSPOT and SomaKit TOC™ have already been approved by the FDA and EMA, respectively [142].
Several approaches for processing generator-derived 68Ga eluates have been recently described, each of them with a markedly different impact in terms of technology and engineering towards the development of automated systems for 68Ga radiolabeling [90]. The product is labeled to the conjugate in question, in a similar manner to that described previously, and the product obtained then purified from any unlabeled 68Ga using a C18 cartridge. The radiopharmaceutical is eluted from the C18 cartridge using ethanol or an ethanol/water mixture and eventually diluted with 0.9% saline as the final product. To ensure sterility, the final product is eluted through a 0.22 μm sterilization filter. Instant thin layer chromatography (ITLC) as well as HPLC are performed as a quality control to ensure the radiochemical purity (RCP) of the product is above 95% and the fraction of free 68Ga+ or its colloids is less than 2%, to allow the release of the product for clinical administration. Germanium breakthrough needs to be assessed with the quantity limited to a maximum of 0.001% [143,144,145].
As it was mentioned in Section 2.4, the precise activity measurements are essential for dosing and absolute image quantification. In a recent study, accuracy of four different types of radionuclide calibrators was investigated for 68Ga by using well-calibrated gamma spectrometry. The results were surprising in that all the radionuclide calibrator systems showed a systematic error in the range of 10–25% [76,146]. It was reported that the deviations were mainly due to the incorrect factory-shipped dose calibrator setting.
At the time of writing, a specification exists only for the generator-produced 68Ga in the European Pharmacopoeia monographs (Table 4), however, a new monograph of the European Pharmacopeia is in preparation for accelerator-produced 68Ga solution [75]. Specifications for generator or cyclotron produced 68Ga are similar except for the radionuclidic purity, which is proposed to be >98% (<2% 66Ga and 67Ga) for cyclotron-produced 68Ga (Table 5).

3.4.2. 44. Sc

There is no monograph in the European or another pharmacopeia available for the preparation of 44Sc or 44Sc-radiopharmaceuticals. To date, the quality control was performed based on the monograph for [68Ga]Ga-DOTATOC of the European Pharmacopeia. As Sc is chemically more similar to the lanthanides than Ga, the labeling procedure would be potentially similar to that of 177Lu. The labeling pH should be taken into consideration as was pointed out in the previous section. Thanks to the promising preclinical studies with 44Sc, proof-of-principle first-in-man studies were performed with [44Sc]Sc-PSMA-617 and 44Sc-DOTATOC [35,149], demonstrating that PET/CT imaging can be performed several hours post-injection (19 and 24 h) thanks to the longer half-life of 44Sc.

4. Beta-Emitting Radionuclides: 161Tb and 177Lu

4.1. Targetry Production

4.1.1. 177Lu

The main methods for producing 177Lu with high specific activity are based on neutron irradiation of either enriched 176Lu or 176Yb in reactors. Lu2O3 or Yb2O3 are used as target material, due to their thermal stability (during irradiation) and solubility in diluted mineral acids [10].

Direct Method: Production of Carrier-Added (c.a.) 177Lu

The direct production of 177Lu takes place via the 176Lu(n,γ)177Lu nuclear reaction. 176Lu has a very high thermal neutron capture cross-section (σ: 2090 b), which allows reaching high yields and relatively high specific activity. The co-production of 177mLu (T1/2 = 160.4 d), however, is an issue for radiation protection and waste disposal (laboratory waste, hospital wastewater). The irradiated material, Lu2O3, is easily dissolved in concentrated hydrochloric acid and heated until dryness to convert the oxide to chloride. The resultant chloride residue is dissolved in dilute hydrochloric acid and it is used directly for labeling. In this phase of preparation, contamination due to the impurities in the initial material (target material or quartz ampoule) and during the processing of the irradiated material, which can directly affect the labeling efficiency, should be taken into consideration.
Based on theoretical calculations, no long-lived radionuclidic impurities, except for 177mLu, are expected to be found in the irradiated lutetium. Barkhausen et al. performed gamma spectrometry measurements three weeks after irradiation, to confirm these data and the obtained results showed the presence of only 169Yb (T1/2: 32.026 d) and 65Zn (T1/2: 244.26 d) as contaminants. However, at the end of irradiation, their relative activities were very low 1.3 × 10−4 and 1.8 × 10−4 %, respectively [150].

Indirect Method: Production of No-Carrier-Added (n.c.a.) 177Lu

The production of n.c.a. 177Lu occurs by the irradiation of enriched 176Yb via 176Yb(n,γ)177Yb→177Lu nuclear reaction. The production yields are much lower compared to the direct method due to the low thermal neutron cross-section of 176Yb (2.5 barn). It requires a well-developed radiochemical separation method to be able to reach high labeling efficiencies and for the recycling of the enriched 176Yb. Enriched 176Yb oxide contains 2–3% of 174Yb, which also leads to the formation of 175Yb (T1/2 = 4.18 d). This method is much more expensive compared to the direct method, however, it ensures production of 177Lu with high radionuclidic purity without the presence of long-lived radioactive impurities, which minimizes the radiation protection and waste disposal issues. In addition, the shelf-life of n.c.a. 177Lu product is much longer compared to c.a. 177Lu.

4.1.2. 161. Tb

Indirect method: production of no-carrier-added 161Tb. The production of n.c.a. 161Tb takes place with the irradiation of enriched 160Gd via the 160Gd(n,γ)161Gd→161Tb nuclear reaction. 159Tb is the only stable isotope of terbium, therefore, a direct production route is not an option to produce 161Tb.
A method to produce 161Tb towards medical application was first proposed by Lehenberger et al. [151]. Table 6 shows the comparison of 177Lu and 161Tb decay characteristics and their relative production routes. The 176Yb thermal neutron reaction cross-section (2.5 barn) is high compared to that of 160Gd (1.5 barn). As a result, longer irradiation times or higher flux are required for the production of the same activities for 161Tb. The natural abundance of 160Gd (21.86%) is higher than 176Yb (12.76%), however, which makes the target material to be irradiated less expensive (Table 7).
Production of 161Tb was also investigated via deuteron-induced reactions (160Gd(d, x)161Tb) over a wide energy range up to 50 MeV, using the stacked-foil technique and high-resolution γ-spectrometry [47]. Nevertheless, it was reported that, even when using 160Gd enriched targets, the radionuclidic purity of the final product cannot be high due to the co-production of 160Tb.
161Tb has not yet been listed on the dangerous goods tables of the ADR (European Agreement concerning the International Carriage of Dangerous Goods by Road) and International Air Transport Agency (IATA) regulations, which are based on IAEA (International Atomic Energy Agency) recommendations. As a result, the generic A2 value (activity limit of radioactive material) according to the “Basic Radionuclide Values for Unknown Radionuclides or Mixtures” of 0.02 TBq has to be applied for 161Tb transportation [154]. Based on the current limitation of irradiation conditions (due to the transportation limit), the specific activity of 161Tb has yet to be optimized.

4.2. Radiochemistry

4.2.1. 177. Lu

Due to their chemical similarity, the separation of neighboring lanthanides is challenging. The effective separation of micro amounts of 177Lu product from macro amounts of Yb target material, additionally, needs special attention since high separation factors (>105) are required. The most common method to separate lanthanides is the use of α-HIBA (α-hydroxyisobutryric acid) as eluent in a cation exchanger separation system [155]. To reach high separation factors, α-HIBA concentration, pH of the solution and the parameters for the cation exchanger (column volume, flow rate, particle size, etc.) should be optimized.
Several methods have been proposed in the literature for the separation of 177Lu from Yb [156,157,158,159,160,161]. Kuznetsov et al., reviewed the separation of Lu and Yb by reduction of Yb with sodium amalgam and subsequent developments [19]. The radiochemical separation method for the routine production of n.c.a. 177Lu on an industrial scale for medical applications is described in [162]. The reported separation method is capable of the separation of 177Lu and 176Yb in a mass ratio of 1:102 to 1:1010. It contains a five cation exchange column system. Firstly, Yb2O3 is dissolved in mineral acid, then, the solution is loaded to the first column system (macroporous cation exchanger in NH4+ form). The first two columns are used to separate the macro amounts of Yb from the micro amount of Lu. Then, the desired separation factor for Lu is reached with further separation of Yb performed by the next two columns. The α-HIBA is solution used as a chelating agent to separate Lu and Yb. After elution of Lu from the second column, the macro amounts Yb is eluted with a higher concentration of HIBA and collected for recycling.
The final Lu solution is acidified to dissociate the α-HIBA-Lu complex and it is eluted from the fifth column in 1.0 M to 12.0 M mineral acid. The mineral acid is then evaporated and the product prepared by collection in the desired solution. The final product is suited for radiopharmaceutical use without requiring further purification and/or sterilization. It was reported that with this method, 3.9 TBq of 177Lu/mg of lutetium could be obtained and more than 400 MBq 177Lu could be labeled per µg peptide or other biomolecules [161].

4.2.2. 161. Tb

A two-column separation system has been applied for the separation of terbium from enriched gadolinium target material [162]. After dissolution of target and the loading of the radionuclides onto a Sykam cation exchange resin column, α-HIBA separation system is used to separate terbium from macro amounts of gadolinium target material.
The α-HIBA-Tb complex must be decomposed before labeling. This compound is very stable and its existence in the final product would hinder the labeling process. LN3 resin is an extraction resin containing acidic organophosphorus extractants [163], which can be used for the separation of complexing agent (α-HIBA) from Tb and, furthermore, ensures elution of 161Tb in a small volume of dilute HCl. The use of LN3 resin, as suggested by Gracheva et al., is used as the last step of 161Tb production to avoid the lengthy evaporation step [162], previously needed to obtain the final solution in dilute acid. This procedure cannot replace the evaporation step in the 177Lu separation process, however, due to the high distribution coefficient of Lu in dilute acids on LN3 resin [164].
When using the α-HIBA separation system, the separation factors for Lu/Yb (1.55) and Gd/Tb (1.57) are very close [165]. In addition, both radionuclides’ production processes require the recovery of the enriched target material. The elemental analysis of the recycled target material is essential to determine the quality of the material before irradiation.

4.3. Labeling Efficiency

Other than their decay properties, another intriguing feature of the radiolanthanides is that they share similar chemical characteristics such as analogous coordination chemistry. They exist mainly in the +3 oxidation state and form eight coordination complexes with smaller ions and nine coordination complexes with larger ions. As a result, they can be linked to a variety of molecular carriers such as small molecules, peptides, proteins and antibodies [166,167,168,169,170].
Lanthanides cations form insoluble hydroxides at pH >6, making the optimal pH for radiolabeling in the range of pH 4-6. An identical labeling procedure can be applied for both 161Tb and 177Lu thanks to the similar coordination chemistry. The decay products of 177Lu and 161Tb are Hf4+ and Dy3+, respectively. Breeman et al. reported that Hf4+ does not interfere with Lu binding with the DOTA-chelator [78]. On the other hand, the decay product of 161Tb is Dy, which could well interfere with Tb labeling. The radiolabeling yield of 161Tb-DOTA has been investigated and it was shown that even after two weeks, the molar activity of the product was sufficient for potential clinical application [162]. Comparison studies have shown that 161Tb- and 177Lu-DOTATOC (solutions in saline at room temperature) are stable over 24 h in the presence of a stabilizer (L-ascorbic acid).

4.4. Production for the Clinical Applications

4.4.1. 177. Lu

Currently, commercial n.c.a. 177Lu is produced and supplied to the market by ITG, Germany [20] and several GMP protocols have been developed for the synthesis of 177Lu-labeled radiopharmaceuticals. ITG has Marketing Authorization and supplies 177Lu under the product name EndolucinBeta. Few studies, using complete automated labeling processes of peptides or antibodies with 177Lu, have been reported to date. Syntheses investigated are mainly DOTA-based derivatives [91,92,143,171,172]. Sodium ascorbate solution is used as a buffer to adjust the reaction pH from 4.5-4.9. The volume of the precursor (DOTATOC or PSMA-617) is taken based on the delivered radioactivity. Then, the buffer/precursor solutions were transferred to the reaction vial and heated (90oC) for 30 min [171]. In these systems, after the synthesis is carried out, the product from the reaction vial is loaded onto a solid phase extraction (SPE) C18 cartridge to purify the final product from any free activity still in the solution. The radiopharmaceutical is then eluted from the C18 cartridge with an ethanol/water mixture and diluted with 0.9% saline into the final product vial. The final product is eluted through a 0.22 μm filter to ensure sterility. In the development stage, instant thin layer chromatography (ITLC), as well as high pressure liquid chromatography (HPLC), are performed as quality control to determine the radiochemical purity of the product. At this stage, sterility and endotoxin are usually not assessed [91,171,172].

4.4.2. 161. Tb

161Tb is not yet commercially available and is not produced based on pharmaceutical legislation in EU-GMP regulations. It was demonstrated that the 161TbCl3 final product is suitable for pharmaceutical use without requiring further purification and sterilization [162]. Due to the similar chemical characteristics of 161Tb and 177Lu, the synthesis module protocols commonly used for 177Lu synthesis can be directly adapted for the production of 161Tb radiopharmaceuticals. Nevertheless, 161TbCl3 solution has to be validated for GMP use.
Moreover, it is worth mentioning that, due to the low-energy gamma photons which lead to substantial self-absorption, the geometry effect on the activity measurement (see Section 2.1) with dose calibrators for 161Tb should be assessed. Table 8 shows that the quality of 161Tb is comparable with commercially-available 177Lu (EndolucinBeta-ITG) and it is suitable for future clinical applications.

5. Conclusions

Radionuclide production is the first step for the preparation of radiopharmaceuticals. To achieve the aim of routine large-scale production, process development of each step in the radionuclide production process is vital. It should be also underlined that radionuclide production for nuclear medical applications is a process under continuous development and optimization.
The success of preclinical studies strongly depends on the availability of the novel radionuclides, as well as the quality thereof. The number of high-intensity intermediate/high-energy and multiple-particle cyclotrons plays a crucial role in the development and the future availability of the novel radionuclides. Furthermore, it is foreseen that in the near future, the sources of nuclear research reactors will not be sufficient due to the wide use of therapeutic β--emitting radionuclides.
161Tb and 44Sc are novel radionuclides for radiotherapy and PET application, respectively, and seen as a possible improvement to what is currently utilized in a clinical setting. Currently, development of these radionuclides has progressed to the point where the radionuclide precursor reproducibly meets the required quality and the protocols being developed for an automated module for the direct radiolabeling. However, up to date, there is no monograph in the European Pharmacopeia for the preparation of 161Tb and 44Sc radiopharmaceuticals and there is an urgent need for the definition of the regulation and legislation of the manufacturing thereof, as well as the related quality documentation of the radionuclide precursors. In parallel, more preclinical and clinical comparison studies with 161Tb and 177Lu will successfully guide the translation of 161Tb to the clinics.

Author Contributions

Every author contributed to this review. All authors have read and agreed to the published version of the manuscript.

Funding

This project was funded by the Swiss National Science Foundation (SNF grant number: 200021_188495).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. European Commission. Supply of medical isotopes. Available online: https://ec.europa.eu/euratom/observatory_radioisotopes.html (accessed on 30 January 2020).
  2. Fior Markets, Global Radiopharmaceuticals/Nuclear Market by Type (Diagnostic, Therapeutic), procedure, Application, End User, Region, Global Industry Analysis, Market Size, Share, Growth, Trends, and Forecast 2018 to 2025. Available online: https://www.fiormarkets.com/report/global-radiopharmaceuticals-nuclear-market-by-type-diagnostic-therapeutic-procedure-376060.html (accessed on 30 January 2020).
  3. Ku, A.; Facca, V.J.; Cai, Z.; Reilly, R.M. Auger electrons for cancer therapy–a review. EJNMMI Radiopharma. Chem. 2019, 4, 1–36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Kassis, A.I. Cancer Therapy with Auger Electrons: Are We Almost There ? J. Nucl. Med. 2003, 44, 1479–1482. [Google Scholar] [PubMed]
  5. Howell, R.W. Auger processes in the 21st century. Int. J. Radiat. Biol. 2008, 84, 959–975. [Google Scholar] [CrossRef] [PubMed]
  6. Kassis, A.I. Molecular and cellular radiobiological effects of Auger emitting radionuclides. Radiat. Prot. Dosimetry 2011, 143, 241–247. [Google Scholar] [CrossRef] [Green Version]
  7. Kassis, A.I. The amazing world of Auger electrons. Int. J. Radiat. Biol. 2004, 80, 789–803. [Google Scholar] [CrossRef]
  8. Ahmadzadehfar, H.; Eppard, E.; Kürpig, S.; Fimmers, R.; Yordanova, A.; Schlenkhoff, C.D.; Gärtner, F.; Rogenhofer, S.; Essler, M. Therapeutic response and side effects of repeated radioligand therapy with 177Lu-PSMA-DKFZ-617 of castrate-resistant metastatic prostate cancer. Oncotarget 2016, 7, 12477–12488. [Google Scholar] [CrossRef]
  9. Rahbar, K.; Ahmadzadehfar, H.; Kratochwil, C.; Haberkorn, U.; Sch, M.; Essler, M.; Baum, R.P.; Kulkarni, H.R.; Schmidt, M.; Drzezga, A.; et al. German Multicenter Study Investigating 177Lu-PSMA-617 Radioligand Therapy in Advanced Prostate Cancer. J. Nucl. Med. 2017, 58, 85–90. [Google Scholar] [CrossRef] [Green Version]
  10. Baum, R.P.; Kulkarni, H.R.; Schuchardt, C.; Singh, A.; Wirtz, M.; Wiessalla, S.; Schottelius, M.; Mueller, D.; Klette, I.; Wester, H.J. 177Lu-labeled prostate-specific membrane antigen radioligand therapy of metastatic castration-resistant prostate cancer: Safety and efficacy. J. Nucl. Med. 2016, 57, 1006–1013. [Google Scholar] [CrossRef] [Green Version]
  11. Pfeifer, A.K.; Gregersen, T.; Gronbaek, H.; Hansen, C.; Brand, J.M.; Bruun, K.H.; Krogh, K.; Kjaer, A.; Knigge, U. Peptide Receptor Radionuclide Therapy with Y-DOTATOC and 177Lu-DOTATOC in Advanced Neuroendocrine Tumors: Results from a Danish Cohort Treated in Switzerland. Neuroendocrinology 2011, 93, 189–196. [Google Scholar] [CrossRef]
  12. Mantry, P.S.; Mehta, A.; Madani, B.; Mejia, A.; Shahin, I. Selective internal radiation therapy using yttrium-90 resin microspheres in patients with unresectable hepatocellular carcinoma: A retrospective study. J. Gastrointest. Oncol. 2017, 8, 799–807. [Google Scholar] [CrossRef] [Green Version]
  13. Furubayashi, N.; Negishi, T.; Ura, S.; Hirai, Y.; Nakamura, M. Palliative effects and adverse events of strontium-89 for prostate cancer patients with bone metastasis. Mol. Clin. Oncol. 2015, 3, 257–263. [Google Scholar] [CrossRef] [PubMed]
  14. Yamada, K.; Yoshimura, M.; Kaise, H.; Ogata, A.; Ueda, N.; Tokuuye, K.; Kohno, N. Concurrent use of Sr-89 chloride with zoledronic acid is safe and effective for breast cancer patients with painful bone metastases. Exp. Ther. Med. 2012, 3, 226–230. [Google Scholar] [CrossRef] [PubMed]
  15. Dionisi, V.; Bellarosa, C.; Cardano, R.; Rizzini, E.L.; Ghedini, P.; Morganti, A.G.; Fanti, S.; Monari, F. Use of Radium-223 Dichloride in Patients With Osteonecrosis of the Jaw Induced by Zoledronic Acid: Report of 2 Cases. Clin. Genitourin. Cancer 2019, 17, e612–e617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Den, R.B.; George, D.; Pieczonka, C.; McNamara, M. Ra-223 treatment for bone metastases in castrate-resistant prostate cancer: Practical management issues for patient selection. Am. J. Clin. Oncol. Cancer Clin. Trials 2019, 42, 399–406. [Google Scholar] [CrossRef] [PubMed]
  17. Carroll, P.R. NCCN Guidelines Insights Prostate cancer early detection, version 2.2016. J. Nat. Compr. Canc. Netw. 2016, 14, 509–519. [Google Scholar] [CrossRef] [PubMed]
  18. Kratochwil, C.; Fendler, W.; Eiber, M.; Baum, R.; Bozkurt, M.F.; Czernin, J.; Bolton, R.C.D.; Ezziddin, S.; Forrer, F.; Hicks, R.J. EANM procedure guidelines for radionuclide therapy with 177Lu-labelled PSMA-ligands ( 177Lu-PSMA-RLT ). Eur. J. Nucl. Med. Mol. Imaging 2019, 46, 2536–2544. [Google Scholar] [CrossRef]
  19. Kuznetsov, R.A.; Bobrovskaya, K.S.; Svetukhin, V.V.; Fomin, A.N.; Zhukov, A. V Production of Lutetium-177: Process Aspects. Radiochemistry 2019, 61, 381–395. [Google Scholar] [CrossRef]
  20. Dash, A. Production of 177Lu for Targeted Radionuclide Therapy: Available Options. Nucl. Med. Mol Imaging 2015, 49, 85–107. [Google Scholar] [CrossRef] [Green Version]
  21. European Medicines Agency, EndolucinBeta International. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/endolucinbeta (accessed on 30 January 2020).
  22. NuDat 2.8, Decay radiation search. Available online: https://www.nndc.bnl.gov/nudat2/ (accessed on 30 January 2020).
  23. Hindie, E.; Zanotti-Fregonara, P.; Quinto, M.A.; Morgat, C.; Champion, C. Dose Deposits from 90Y, 177Lu, 111In, and 161Tb in Micrometastases of Various Sizes: Implications for Radiopharmaceutical Therapy. J. Nucl. Med. 2016, 57, 759–764. [Google Scholar] [CrossRef] [Green Version]
  24. Champion, C.; Quinto, M.A.; Morgat, C.; Zanotti-fregonara, P.; Hindié, E. Comparison between Three Promising ß-emitting Radionuclides, 67Cu, 47Sc and 161Tb, with Emphasis on Doses Delivered to Minimal Residual Disease. Theranostics 2016, 6, 1611–1618. [Google Scholar] [CrossRef] [Green Version]
  25. Müller, C.; Reber, J.; Haller, S.; Dorrer, H.; Bernhardt, P.; Zhernosekov, K.; Türler, A.; Schibli, R. Direct in vitro and in vivo comparison of 161Tb and 177Lu using a tumour-targeting folate conjugate. Eur J. Nucl. Med. Mol Imaging 2014, 41, 476–485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Müller, C.; Umbricht, C.A.; Gracheva, N.; Tschan, V.J.; Pellegrini, G.; Bernhardt, P.; Zeevaart, J.R.; Köster, U.; Schibli, R.; Meulen, N.P. Van Der Terbium-161 for PSMA-targeted radionuclide therapy of prostate cancer. Eur. J. Nucl. Med. Mol. Imaging 2019, 46, 1919–1930. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Grünberg, J.; Lindenblatt, D.; Dorrer, H.; Cohrs, S.; Zhernosekov, K.; Köster, U. Anti-L1CAM radioimmunotherapy is more effective with the radiolanthanide terbium-161 compared to lutetium-177 in an ovarian cancer model. Eur J. Nucl. Med. Mol Imaging 2014, 41, 1907–1915. [Google Scholar] [CrossRef] [PubMed]
  28. Marin, I.; Ryden, T.; van Essen, M.; Svensson, J.; van der Meulen, N.P.; Gracheva, N.; Müller, C. Establishment of optimized SPECT/CT protocol for therapy with 161Tb demonstrate improved image quality with Monte Carlo based OSEM reconstruction. Eur J. Nucl. Med. Mol Imaging 2018, 45, S16. [Google Scholar]
  29. Müller, C.; Domnanich, K.A.; Umricht, C.A.; Van der Meulen, N. Theranostics and precision medicine special feature: Review Article Scandium and terbium radionuclides for radiotheranostics: Current state of development towards clinical application. Br. J. Radiol 2018, 91, 1–13. [Google Scholar] [CrossRef] [PubMed]
  30. Qaim, S.M.; Scholten, B.; Neumaier, B. New developments in the production of theranostic pairs of radionuclides. J. Radioanal. Nucl. Chem. 2018, 318, 1493–1509. [Google Scholar] [CrossRef] [Green Version]
  31. Domnanich, K.A.; Müller, C.; Benešová, M.; Dressler, R.; Haller, S.; Köster, U.; Ponsard, B.; Schibli, R.; Türler, A.; van der Meulen, N.P. 47Sc as useful β–-emitter for the radiotheragnostic paradigm: A comparative study of feasible production routes. EJNMMI Radiopharm. Chem. 2017, 2, 1–17. [Google Scholar] [CrossRef] [Green Version]
  32. Müller, C.; Bunka, M.; Haller, S.; Köster, U.; Groehn, V.; Bernhardt, P.; Van Der Meulen, N.; Türler, A.; Schibli, R. Promising prospects for 44Sc-/47Sc-based theragnostics: Application of 47Sc for radionuclide tumor therapy in mice. J. Nucl. Med. 2014, 55, 1658–1664. [Google Scholar] [CrossRef] [Green Version]
  33. Siwowska, K.; Guzik, P.; Domnanich, K.A.; Rodríguez, J.M.M.; Bernhardt, P.; Ponsard, B.; Hasler, R.; Borgna, F.; Schibli, R.; Köster, U.; et al. Therapeutic potential of 47Sc in comparison to 177Lu and 90Y: Preclinical investigations. Pharmaceutics 2019, 11, 424. [Google Scholar] [CrossRef] [Green Version]
  34. Umbricht, C.A.; Benesova, M.; Schmid, R.M.; Türler, A.; Schibli, R.; van der Meulen, N.P.; Müller, C. Sc-PSMA-617 for radiotheragnostics in tandem with 177Lu-PSMA-617 — preclinical investigations in comparison with. EJNMMI Res. 2017, 7, 1–10. [Google Scholar] [CrossRef] [Green Version]
  35. Eppard, E.; de Fuente, A.; Benešová, M.; Khawar, A.; Bundschuh, R.A.; Gärtner, C.; Kreppel, B.; Kopka, K.; Essler, M.; Rösch, F. Clinical Translation and First In-Human Use of [ 44Sc ] Sc-PSMA-617 for PET Imaging of Metastasized Castrate-Resistant Prostate Cancer. Theranostics 2017, 7, 4359–4369. [Google Scholar] [CrossRef] [PubMed]
  36. Bunka, M.; Müller, C.; Vermeulen, C.; Haller, S.; Türler, A.; Schibli, R.; Meulen, N.P. van der Imaging quality of 44Sc in comparison with five other PET radionuclides using Derenzo phantoms and preclinical PET. Appl. Radiat. Isot. 2016, 110, 129–133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. García-Toraño, E.; Peyrés, V.; Roteta, M.; Sánchez-Cabezudo, A.I.; Romero, E.; Martínez Ortega, A. Standardization and precise determination of the half-life of 44Sc. Appl. Radiat. Isot. 2016, 115, 313. [Google Scholar] [CrossRef] [PubMed]
  38. Lee, H.H.; Chen, J.C. Investigation of attenuation correction for small-animal single photon emission computed tomography. Comput. Math. Methods Med. 2013, 2013, 1–13. [Google Scholar] [CrossRef]
  39. Stolarz, A. Target preparation for research with charged projectiles. J. Radioanal Nucl. Chem 2014, 299, 913–931. [Google Scholar] [CrossRef] [Green Version]
  40. Skliarova, H.; Cisternino, S.; Cicoria, G.; Marengo, M.; Cazzola, E.; Gorgoni, G.; Palmieri, V. Medical Cyclotron Solid Target Preparation by Ultrathick Film Magnetron Sputtering Deposition. Instruments 2019, 3, 21. [Google Scholar] [CrossRef] [Green Version]
  41. SRIM. Stopping and Range of Ions in matter. Available online: http://www.srim.org/ (accessed on 30 January 2020).
  42. Alves, V.; do Carmo, S.; Alves, F.; Abrunhosa, A. Automated Purification of Radiometals Produced by Liquid Targets. Instruments 2018, 2, 17. [Google Scholar] [CrossRef] [Green Version]
  43. Do Carmo, S.J.C.; Scott, P.J.H.; Alves, F. Production of radiometals in liquid targets. EJNMMI Radiopharm. Chem. 2020, 5, 1–21. [Google Scholar] [CrossRef]
  44. Pandey, M.K.; Engelbrecht, H.P.; Byrne, J.P.; Packard, A.B.; DeGrado, T.R. Production of 89Zr via the 89Y(p,n)89Zr reaction in aqueous solution: Effect of solution composition on in-target chemistry. Nucl. Med. Biol. 2014, 41, 309–316. [Google Scholar] [CrossRef]
  45. Pawlak, D.; Parus, J.L.; Sasinowska, I.; Mikolajczak, R. Determination of elemental and radionuclidic impurities in 177Lu used for labeling of radiopharmaceuticals. J. Radioanal. Nucl. Chem. 2004, 261, 469–472. [Google Scholar] [CrossRef]
  46. Qaim, S.M. Nuclear data for production and medical application of radionuclides: Present status and future needs. Nucl. Med. Biol. 2017, 44, 31–49. [Google Scholar] [CrossRef] [PubMed]
  47. Tarkanyi, F.; Hermanne, A.; Ditroi, F.; Csikai, J.; Ignatyuk, A.V. Cross-section measurement of some deuteron induced reactions on 160Gd for possible production of the therapeutic radionuclide 161Tb. J. Radioanal Nucl Chem 2013, 298, 1385–1392. [Google Scholar] [CrossRef] [Green Version]
  48. Engle, J.W.; Ignatyuk, A.V.; Capote, R.; Carlson, B.V.; Hermanne, A.; Kellett, M.A.; Kib, T. Recommended Nuclear Data for the Production of Selected Therapeutic Radionuclides. Nucl. Data Sheets 2019, 155, 56–74. [Google Scholar] [CrossRef]
  49. Tarkanyi, F.T.; Ignatyuk, A.V.; Hermanne, A.; Capote, R.; Carlson, B.V.; Engle, J.W.; Kellett, M.; Kibedi, M.; Kondev, G.; Lebeda, M.; et al. Recommended nuclear data for medical radioisotope production: Diagnostic gamma emitters. J. Radioanal. Nucl. Chem. 2019, 319, 487–531. [Google Scholar] [CrossRef] [Green Version]
  50. Tarkanyi, F.T.; Ignatyuk, A.V.; Hermanne, A.; Engle, J.W.; Kellett, M.A.; Kibedi, T.; Kim, G.; Kondev, F.; Lebeda, O.; Verpelli, M. Recommended nuclear data for medical radioisotope production: Diagnostic positron emitters. J. Radioanal. Nucl. Chem. 2019, 319, 533–666. [Google Scholar] [CrossRef] [Green Version]
  51. Sajjad, M.; Lambrecht, M. Cyclotron targetry for medical radioisotope production. Nucl. Instruments Methods Phys. Res. B 1989, 41, 1100–1104. [Google Scholar] [CrossRef]
  52. Otuka, N.; Takács, S. Definitions of radioisotope thick target yields. Radiochim. Acta 2015, 103, 1–6. [Google Scholar] [CrossRef] [Green Version]
  53. Goethals, P.E.; Zimmermann, R. Cyclotrons used in Nuclear Medicine World Market. Report Directory. 2015. Available online: http://medraysintell.com/attachments/File/MEDraysintell_Cyclotron_Edition_2015_-_Summary_and_TOC.pdf (accessed on 30 January 2020).
  54. van der Meulen, N.P.; Eichler, R.; Grundler, P.V.; Hasler, R.; Hirzel, W.; Joray, S.; Kiselev, D.C.; Sommerhalder, A.; Talip, Z.; Zhang, H.; et al. The use of PSI’s IP2 beam line towards exotic radionuclide development and its application towards proof-of-principle preclinicaland clinical studies. In Proceedings of the 22th International Conference on Cyclotrons and their Applications, National Reseach Foundation, Cape Town, South Africa, 22–27 September 2019; p. 24. [Google Scholar]
  55. IAEA. Research Reactor Database. Available online: https://nucleus.iaea.org/RRDB/Content/Util/IsoTopes.aspx (accessed on 30 January 2020).
  56. Krijger, G.C.; Ponsard, B.; Harfensteller, M.; Wolterbeek, H.T.; Nijsen, J.W.F. The necessity of nuclear reactors for targeted radionuclide therapies. Trends Biotechnol. 2013, 31, 390–396. [Google Scholar] [CrossRef]
  57. Fedosseev, V.; Marsh, B.; Fedorov, D.V.; Koester, U. Ionization scheme development at the ISOLDE MLIS. Hyperfine Interact. 2005, 3, 1–13. [Google Scholar]
  58. Fedosseev, V.; Chrysalidis, K.; Goodacre, T.D.; Marsh, B.; Rothe, S.; Seiffert, C.; Wendt, K. Ion beam production and study of radioactive isotopes with the laser ion source at ISOLDE Ion. J. Phys. G Nucl. Part. Phys. 2017, 44, 1–26. [Google Scholar] [CrossRef]
  59. Santos Augusto, R.; Stora, T.; Buehler, L.; Lawson, Z.; Marzari, S.; Stachura, M.; Stora, T.; Collaboration, C.-M. CERN-MEDICIS (Medical Isotopes Collected from ISOLDE): A New Facility. Appl. Sci. 2014, 4, 265–281. [Google Scholar] [CrossRef]
  60. Gadelshin, V.; Cocolios, T.; Fedoseev, V.; Heinke, R.; Kieck, T.; March, B.; Naubereit, P.; Rothe, S.; Stora, T.; Studer, D.; et al. Laser resonance ionization spectroscopy on lutetium for the MEDICIS project. Hyperfine Interact. 2017, 238, 1–7. [Google Scholar] [CrossRef] [Green Version]
  61. Köster, U.; Assmann, W.; Bacri, C.; Faestermann, T.; Garrett, P.; Gernhäuser, R.; Tomandl, I. Electromagnetic isotope separation of gadolinium isotopes for the production of 152,155Tb for radiopharmaceutical applications. Nucl. Inst. Methods Phys. Res. B 2020, 463, 111–114. [Google Scholar] [CrossRef]
  62. Hoehr, C.; Bénard, F.; Buckley, K.; Crawford, J.; Gottberg, A.; Hanemaayer, V. Medical isotope production at TRIUMF – from imaging to treatment. Phys. Procedia 2017, 90, 200–208. [Google Scholar] [CrossRef]
  63. Baeten, P.; Schyns, M.; Fernandez, R.; de Bruyn, D.; Eynde, G. Van Den MYRRHA: A multipurpose nuclear research facility. In Proceedings of the EPJ Web of Conferences, Rhodes, Greece, 25–30 August 2013; EDP Sciences: Les Ulis, France, 2014; pp. 1–7. [Google Scholar]
  64. Formento-Cavaier, R.; Köster, U.; Crepieux, B.; Gadelshin, V.M.; Haddad, F.; Stora, T.; Wendt, K. Very high specific activity erbium 169Er production for potential receptor-targeted radiotherapy. Nucl. Instruments Methods Phys. Res. Sect. B Beam Interact. Mater. Atoms 2019, 463, 468–471. [Google Scholar] [CrossRef]
  65. Starovoitova, V.N.; Tchelidze, L.; Wells, D.P. Production of medical radioisotopes with linear accelerators. Appl. Radiat. Isot. 2014, 85, 39–44. [Google Scholar] [CrossRef]
  66. Jang, J.; Yamamoto, M.; Uesaka, M. Design of an X-band electron linear accelerator dedicated to decentralized 99Mo/99mTc supply: From beam energy selection to yield estimation. Phys. Rev. Accel. Beams 2017, 20, 104701–104709. [Google Scholar] [CrossRef]
  67. Jones, S.; JR Robinson, G.D.; Mcintyre, E. Tandem Van de Graaff Accelerator Production of Positron Labeled Radiopharmaceuticals for Routine Clinical Use. Int. J. Appl. Rdiat. Hot. Vol. 1984, 35, 721–729. [Google Scholar] [CrossRef]
  68. Bychenkov, V.Y.; Brantov, A.V.; Mourou, G. Tc-99m production with ultrashort intense laser pulses. Laser Part. Beams 2014, 32, 605–611. [Google Scholar] [CrossRef]
  69. Ledingham, K.; Mckenna, P.; Mccanny, T. High power laser production of short-lived isotopes for positron emission tomography. J. Phys. D Appl. Phys. 2004, 37, 2341–2345. [Google Scholar] [CrossRef] [Green Version]
  70. Lefebvre, E.D.; Humiere, E.; Fritzler, S.; Malka, V. Numerical simulation of isotope production for positron emission tomography with laser- accelerated ions. J. Appl. Phys. 2006, 100, 113308–113318. [Google Scholar] [CrossRef]
  71. Klug, J.; Buckley, K.R.; Zeisler, S.K.; Dodd, M.; Tsao, P.; Hoehr, C.; Economou, C.; Corsaut, J.; Appiah, J.P.; Kovacs, M.S.; et al. A New Transfer System for Solid Targets. In Proceedings of the 14th International Workshop on Targetry and Target Chemistry, Playa Del Carmen, Mexico, 26–29 August 2012; Volume 1509, pp. 146–151. [Google Scholar]
  72. Coenen, H.H.; Gee, A.D.; Adam, M.; Antoni, G.; Cutler, C.S.; Fujibayashi, Y.; Min, J.; Mach, R.H.; Mindt, T.L.; Pike, V.W.; et al. Consensus nomenclature rules for radiopharmaceutical chemistry — Setting the record straight. Nucl. Med. Biol. 2017, 55, 5–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. International Atomic Energy Agency. Quality Assurance for Radioactivity Measurement in Nuclear Medicine; Technical report series no. 454; IAEA: Vienna, Austria, 2006; p. 81. [Google Scholar]
  74. Carey, J.E.; Byrne, P.; De, W.L.; Lieto, R. The Selection, Use, Calibration, and Quality Assurance of Radionuclide Calibrators Used in Nuclear Medicine; Report No: 181; Americal Association of Physicists in Medicine: Alexandria, VA, USA, 2012; p. 38. [Google Scholar]
  75. Council of Europe, European Pharmacopoeia. Available online: https://www.edqm.eu/en/european-pharmacopoeia-ph-eur-9th-edition (accessed on 30 January 2020).
  76. Bauwens, M.; Pooters, I.; Cobben, R.; Visser, M.; Schnerr, R.; Mottaghy, F.; Wildberger, J.; Wierts, R. A comparison of four radionuclide dose calibrators using various radionuclides and measurement geometries clinically used in nuclear medicine. Phys. Medica 2019, 60, 14–21. [Google Scholar] [CrossRef] [PubMed]
  77. Bochud, F.O.; Laedermann, J.P.; Baechler, S.; Kosinski, M.; Bailat, C.J. Usefulness of specific calibration coefficients for gamma-emitting sources measured by radionuclide calibrators in nuclear medicine. Med. Phys. 2011, 38, 4073–4080. [Google Scholar] [CrossRef]
  78. Breeman, W.A.P.; de Jong, M.; Visser, T.J.; Erion, J.L.; Krenning, E.P. Occasional survey Optimising conditions for radiolabelling of DOTA-peptides with 90Y, 111In and 177Lu at high specific activities. Eur J. Nucl Med. Mol Imaging 2003, 30, 917–920. [Google Scholar] [CrossRef]
  79. Asti, M.; Tegoni, M.; Farioli, D.; Iori, M.; Guidotti, C.; Cutler, C.S.; Mayer, P.; Versari, A.; Salvo, D. Influence of cations on the complexation yield of DOTATATE with yttrium and lutetium: A perspective study for enhancing the 90Y and Lu labeling conditions. Nucl. Med. Biol. 2012, 39, 509–517. [Google Scholar] [CrossRef]
  80. Chakravarty, R.; Chakraborty, S.; Dash, A.; Pillai, M.R.A. Detailed evaluation on the effect of metal ion impurities on complexation of generator eluted 68Ga with different bifunctional chelators. Nucl. Med. Biol. 2013, 40, 197–205. [Google Scholar] [CrossRef]
  81. Eppard, E.; Pèrez-malo, M.; Rösch, F. Improved radiolabeling of DOTATOC with trivalent radiometals for clinical application by addition of ethanol. EJNMMI Radiopharm. Chem. 2016, 6, 1–13. [Google Scholar] [CrossRef] [Green Version]
  82. European Pharmacopoeia 10.0. Radiopharmaceutical Preparations; European Directorate for the Quality of Medicines & HealthCare: Strasbourg, France, 2016; pp. 884–887. [Google Scholar]
  83. Pharmaceutical Legistration, EudraLex Volume 4, Good manufacturing practice (GMP) Guidelines. Available online: http://www.it-asso.com/gxp/eudralex_v27/contents/homev4.htm (accessed on 30 January 2020).
  84. EudraLex. The Rules Governing Medicinal Products in the European Union. In Volume 4 Good Manufacturing Practice Medicinal Products for Human and Veterinary Use; European Commission: Brussels, Belgium, 2012; pp. 1–9. [Google Scholar]
  85. Awasthi, V.; Watson, J.; Gali, H.; Matlock, G.; McFarland, A.; Bailey, J.; Anzellotti, A. A “dose on demand” Biomarker Generator for automated production of [18F]F-and [18F]FDG. Appl. Radiat. Isot. 2014, 89, 167–175. [Google Scholar] [CrossRef]
  86. Philippe, C.; Mairinger, S.; Pichler, V.; Stanek, J.; Nics, L.; Mitterhauser, M.; Hacker, M.; Wanek, T.; Langer, O.; Wadsak, W. Comparison of fully-automated radiosyntheses of [11C]erlotinib for preclinical and clinical use starting from in target produced [11C]CO2 or [11C]CH4. EJNMMI Radiopharm. Chem. 2018, 3, 8. [Google Scholar] [CrossRef] [Green Version]
  87. Schultz, M.K.; Mueller, D.; Baum, R.P.; Leonard Watkins, G.; Breeman, W.A.P. A new automated NaCl based robust method for routine production of gallium-68 labeled peptides. Appl. Radiat. Isot. 2013, 76, 46–54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Morley, T.J.; Dodd, M.; Gagnon, K.; Hanemaayer, V.; Wilson, J.; McQuarrie, S.A.; English, W.; Ruth, T.J.; Bénard, F.; Schaffer, P. An automated module for the separation and purification of cyclotron-produced 99mTcO4-. Nucl. Med. Biol. 2012, 39, 551–559. [Google Scholar] [CrossRef] [PubMed]
  89. Elsinga, P.; Todde, S.; Penuelas, I.; Meyer, G.; Farstad, B.; Faivre-Chauvet, A.; Mikolajczak, R.; Westera, G.; Gmeiner-Stopar, T.; Decristoforo, C. Guidance on current good radiopharmacy practice (cGRPP) for the small-scale preparation of radiopharmaceuticals. Eur. J. Nucl. Med. Mol. Imaging 2010, 37, 1049–1062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Boschi, S.; Lodi, F.; Malizia, C.; Cicoria, G.; Marengo, M. Automation synthesis modules review. Appl. Radiat. Isot. 2013, 76, 38–45. [Google Scholar] [CrossRef] [PubMed]
  91. Petrik, M.; Knetsch, P.A.; Knopp, R.; Imperato, G.; Ocak, M.; Von Guggenberg, E.; Haubner, R.; Silbernagl, R.; Decristoforo, C. Radiolabelling of peptides for PET, SPECT and therapeutic applications using a fully automated disposable cassette system. Nucl. Med. Commun. 2011, 32, 887–895. [Google Scholar] [CrossRef]
  92. Ellison, D.; Kaufman, J.; Mather, S.J. Automated radiolabelling of monoclonal antibodies with the Modular Lab system. Nucl. Med. Commun. 2010, 31, 173–177. [Google Scholar] [CrossRef]
  93. Jensen, M.; Clark, J. Direct Production of Ga68 from bombardment of concentrated aqueous solutions of [Zn68] Zicn Chloride. In Proceedings of the 13th International Workshop on Targetry and Target Chemistry; Riso National Laboratory for Sustainable Energy: Roskilde, Denmark, 2011; pp. 288–290. [Google Scholar]
  94. Hoehr, C.; Oehlke, E.; Benard, F.; Lee, C.L.; Hou, X.; Badesso, B.; Ferguson, S.; Miao, Q.; Yang, H.; Buckley, K.; et al. 44gSc production using a water target on a 13 MeV cyclotron. Nucl. Med. Biol. 2014, 41, 401–406. [Google Scholar] [CrossRef]
  95. Pandey, M.K.; Byrne, J.F.; Jiang, H.; Packard, A.B.; Degrado, T.R. Cyclotron production of 68Ga via the 68Zn(p,n)68Ga reaction in aqueous solution. Am. J. Nucl Med. Mol Imaging 2014, 4, 303–310. [Google Scholar]
  96. Pandey, M.K.; Byrne, J.F.; Schlasner, K.N.; Schmit, N.R.; DeGrado, T.R. Cyclotron production of 68Ga in a liquid target: Effects of solution composition and irradiation parameters. Nucl. Med. Biol. 2019, 74–75, 49–55. [Google Scholar] [CrossRef]
  97. Oehlke, E.; Hoehr, C.; Hou, X.; Hanemaayer, V.; Zeisler, S.; Adam, M.J.; Ruth, T.J.; Celler, A.; Buckley, K.; Benard, F.; et al. Production of Y-86 and other radiometals for research purposes using a solution target system. Nucl. Med. Biol. 2015, 42, 842–849. [Google Scholar] [CrossRef]
  98. Abbasi, A.A.; Easwaramoorthy, B. Method and System for Producing Gallium-68 Radioisotope by Solid Targeting in a Cyclotro. Patent WO/2016/197084, 2016. Available online: https://patentscope2.wipo.int/search/en/detail.jsf?docId=WO2016197084 (accessed on 30 January 2020).
  99. van der Meulen, N.P.; Bunka, M.; Domnanich, K.A.; Müller, C.; Haller, S.; Vermeulen, C.; Türler, A.; Schibli, R. Cyclotron production of 44Sc: From bench to bedside. Nucl. Med. Biol. 2015, 42, 745–751. [Google Scholar] [CrossRef] [PubMed]
  100. Krajewski, B.S.; Cydzik, I.; Abbas, K.; Bulgheroni, A.; Simonelli, F.; Holzwarth, U.; Bilewicz, A. Cyclotron production of 44Sc for clinical application. Radiochim. Acta 2013, 101, 333–338. [Google Scholar] [CrossRef]
  101. Engle, J.W.; Lopez-Rodriguez, V.; Gaspar-Carcamo, R.E.; Valdovinos, H.F.; Gonzalez, M.V.-; Trejo-Ballado, F.; Severin, G.W.; Barnhart, T.E.; Nickles, R.J.; Rodriguez, M.A.A.-. Very high specific activity 66/68Ga from zinc targets for PET. Appl Radiat Isot 2012, 70, 1792–1796. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Sadeghi, M.; Kakavand, T.; Rajabifar, S.; Mokhtari, L.; Rahimi-Nezhad, A. Cyclotron production of 68Ga via proton-induced reaction on 68Zn target. Nukleonika 2009, 54, 25–28. [Google Scholar]
  103. Lin, M.; Waligorski, G.J.; Lepera, C.G. Production of curie quantities of 68Ga with a medical cyclotron via the 68Zn (p,n)68Ga reaction. Appl. Radiat. Isot. 2018, 133, 1–3. [Google Scholar] [CrossRef] [PubMed]
  104. Laboratory of Radiochemistry. Annual Report, CaO target development for 44Sc Production; Paul Scherrer Institute: Villigen, Switzerland, 2017; p. 58. [Google Scholar]
  105. Chakravarty, R.; Chakraborty, S.; Ram, R.; Vatsa, R.; Bhusari, P.; Shukla, J.; Mittal, B.R.; Dash, A. Detailed evaluation of different 68Ge/68Ga generators: an attempt toward achieving efficient 68Ga radiopharmacy. J. Label Compd. Radiopharm. 2015, 59, 87–94. [Google Scholar] [CrossRef]
  106. Van der Meulen, N.P.; Dolley, S.G.; Steyn, G.F.; van der Walt, T.N.; Raubenheimer, H.G. The use of selective volatilization in the separation of 68Ge from irradiated Ga targets. Appl. Radiat. Isot. 2011, 69, 727–731. [Google Scholar] [CrossRef]
  107. Rösch, F. Maturation of a Key Resource – The Germanium-68/Gallium-68 Generator: Development and New Insights Maturation of a Key Resource – The Germanium-68/Gallium-68 Generator: Development and New Insights. Curr. Radiopharm. 2012, 5, 202–211. [Google Scholar] [CrossRef]
  108. Alnahwi, A.; Tremblay, S.; Ait-mohand, S.; Beaudoin, J.-F.; Guerin, B. Large-scale routine production of 68Ga using 68Zn-pressed target. J. Nucl. Med. 2019, 60, 634. [Google Scholar]
  109. Filosofov, B.D.V.; Loktionova, N.S.; Rösch, F. A 44Ti/44Sc radionuclide generator for potential application of 44Sc-based PET-radiopharmaceuticals. Radiochim. Acta 2010, 98, 149–156. [Google Scholar] [CrossRef]
  110. Pruszyn’ ski, M.; Loktionova, N.S.; Filosofov, D.V.; Roesch, F. Post-elution processing of Ti-44/Sc-44 generator-derived Sc-44 for clinical application. Appl. Radiat. Isot. 2010, 68, 1636–1641. [Google Scholar] [CrossRef] [PubMed]
  111. Yug, A.; Novgorodov, A.F.; Skripnik, A.V.; D.V., F.; Skripnik, A.V.; Kaplun, V.G.; Suzikov, A.G.; Eliseev, I.A.; Rösch, F. 44Ti: Investigation of target preparation, irradiation and yields in the 45Sc ( p, 2n ) process. Annu. Report. Inst. Nucl. Chem. Univ. Mainz. Available online: https://www.blogs.uni-mainz.de/fb09-kernchemie/files/2018/07/b3_05.pdf (accessed on 30 January 2020).
  112. Zaitseva, N.G.; Tchikalov, M.B.; Khalkin, V.A.; Rurarz, E.; Vobecky, M.; Popinenkova, L.M. Production Cross Sections and Yields of Long Lived 44Ti from 100 MeV Proton Bombardment of Vanadium. Radiochim. Acta 1994, 65, 157–160. [Google Scholar] [CrossRef]
  113. Sajjad, M.; Lambrecht, R.M. Separation of Tracer Titanium-44 from Vanadium. Anal. Chem. 1986, 58, 667–668. [Google Scholar] [CrossRef]
  114. Brodzinski, R.L.; Rancitelli, L.A.; Cooper, J.A.; Wogman, N.A. High-energy proton spallation of iron. Phys. Rev. C 1971, 4, 1257–1265. [Google Scholar] [CrossRef]
  115. Dash, A.; Chakravarty, R. Radionuclide generators: The prospect of availing PET radiotracers to meet current clinical needs and future research demands. Am. J. Nucl Med. Mol Imaging 2019, 9, 30–66. [Google Scholar] [PubMed]
  116. Rösch, F. Scandium-44: Benefits of a Long-Lived PET Radionuclide Available from the 44Ti/44Sc Generator System. Curr. Radiopharm. 2012, 5, 187–201. [Google Scholar] [CrossRef]
  117. Severin, G.W.; Engle, J.W.; Valdovinos, H.F.; Barnhart, T.E.; Nickles, R.J. Cyclotron produced Sc from natural calcium. Appl. Radiat. Isot. 2012, 70, 1526–1530. [Google Scholar] [CrossRef] [Green Version]
  118. Valdovinos, H.F.; Hernandez, R.; Barnhart, T.E.; Graves, S.; Cai, W.; Nickles, R.J. Separation of cyclotron-produced 44Sc from a natural calcium target using a dipentyl pentylphosphonate functionalized extraction resin. Appl Radiat Isot. 2016, 95, 23–29. [Google Scholar] [CrossRef] [Green Version]
  119. Duchemin, C.; Guertin, A.; Haddad, F.; Michel, N.; Métivier, V. Erratum: Production of scandium-44m and scandium-44g with deuterons on calcium-44: Cross section measurements and production yield calculations. Phys. Med. Biol. 2015, 60, 6847–6864. [Google Scholar] [CrossRef]
  120. Alliot, C.; Kerdjoudj, R.; Michel, N.; Haddad, F.; Huclier-Markai, S. Cyclotron production of high purity 44m,44Sc with deuterons from 44CaCO3 targets. Nucl. Med. Biol. 2015, 42, 524–529. [Google Scholar] [CrossRef]
  121. Szkliniarz, K.; Sitarz, M.; Walczak, R.; Jastrzębski, J.; Bilewicz, A.; Choiński, J.; Jakubowski, A.; Majkowska, A.; Stolarz, A.; Trzcińska, A.; et al. Production of medical Sc radioisotopes with an alpha particle beam. Appl. Radiat. Isot. 2016, 118, 182–189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Dash, A.; Chakravarty, R. Pivotal role of separation chemistry in the development of radionuclide generators to meet. RSC Adv. 2014, 4, 42779–42803. [Google Scholar] [CrossRef]
  123. Asti, M.; De Pietri, G.; Fraternali, A.; Grassi, E.; Sghedoni, R.; Fioroni, F.; Roesch, F.; Versari, A.; Salvo, D. Validation of 68Ge/68Ga generator processing by chemical purification for routine clinical application of 68Ga-DOTATOC. Nucl. Med. Biol. 2008, 35, 721–724. [Google Scholar] [CrossRef] [PubMed]
  124. Meyer, G.J.; Mäcke, H.; Schuhmacher, J.; Knapp, W.H.; Hofmann, M. 68Ga-labelled DOTA-derivatised peptide ligands. Eur. J. Nucl. Med. Mol. Imaging 2004, 31, 1097–1104. [Google Scholar] [CrossRef]
  125. Lewis, M.R.; Reichert, D.E.; Laforest, R.; Margenau, W.H.; Shefer, R.E.; Klinkowstein, R.E.; Hughey, B.J.; Welch, M.J. Production and purification of gallium-66 for preparation of tumor-targeting radiopharmaceuticals. Nucl. Med. Biol. 2002, 29, 701–706. [Google Scholar] [CrossRef]
  126. Ugur, Ö.; Kothari, P.J.; Finn, R.D.; Zanzonico, P.; Ruan, S.; Guenther, I.; Maecke, H.R.; Larson, S.M. Ga-66 labeled somatostatin analogue DOTA-DPhe1-Tyr3-octreotide as a potential agent for positron emission tomography imaging and receptor mediated internal radiotherapy of somatostatin receptor positive tumors. Nucl. Med. Biol. 2002, 29, 147–157. [Google Scholar] [CrossRef]
  127. Tolmachew, V.; Lundqvist, H. Rapid Separation of Gallium from Zinc Targets by Thermal Diffusion. Appl. Radiat. Isot. 1996, 47, 297–299. [Google Scholar] [CrossRef]
  128. Sadeghi, M.; Mokhtari, L. Rapid separation of 67,68Ga from 68Zn target using precipitation technique. J. Radioanal. Nucl. Chem. 2010, 284, 471–473. [Google Scholar] [CrossRef]
  129. Wittwer, D.; Dressler, R.; Eichler, R.; Gäggeler, H.W.; Piguet, D.; Serov, A.; Türler, A.; Vögele, A. The thermal release of scandium from titanium metal-A simple way to produce pure 44Sc for PET application. Radiochim. Acta 2011, 99, 193–196. [Google Scholar] [CrossRef] [Green Version]
  130. Zhernosekov, K.P.; Filosofov, D.V.; Baum, R.P.; Aschoff, P.; Bihl, H.; Razbash, A.A.; Jahn, M.; Jennewein, M.; Rösch, F. Processing of generator-produced 68Ga for medical application. J. Nucl. Med. 2007, 48, 1741–1748. [Google Scholar] [CrossRef]
  131. Notni, J.; Wester, H.J. Re-thinking the role of radiometal isotopes: Towards a future concept for theranostic radiopharmaceuticals. J. Label. Compd. Radiopharm. 2017, 61, 141–153. [Google Scholar] [CrossRef] [PubMed]
  132. Ma, M.T.; Cullinane, C.; Waldeck, K.; Roselt, P.; Hicks, R.J.; Blower, P.J. Rapid kit-based 68Ga-labelling and PET imaging with THP-Tyr3-octreotate: A preliminary comparison with DOTA-Tyr3-octreotate. EJNMMI Res. 2015, 5, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Sinnes, J.P.; Nagel, J.; Waldron, B.P.; Maina, T.; Nock, B.A.; Bergmann, R.K.; Ullrich, M.; Pietzsch, J.; Bachmann, M.; Baum, R.P.; et al. Instant kit preparation of 68Ga-radiopharmaceuticals via the hybrid chelator DATA: Clinical translation of [68Ga]Ga-DATA-TOC. EJNMMI Res. 2019, 9, 1–11. [Google Scholar] [CrossRef] [PubMed]
  134. Pruszynski, M.; Majkowska-Pilip, A.; Loktionova, N.S.; Eppard, E.; Roesch, F. Radiolabeling of DOTATOC with the long-lived positron emitter 44Sc. Appl. Radiat. Isot. 2012, 70, 974–979. [Google Scholar] [CrossRef]
  135. Huclier-Markai, S.; Kerdjoudj, R.; Alliot, C.; Bonraisin, A.C.; Michel, N.; Haddad, F.; Barbet, J. Optimization of reaction conditions for the radiolabeling of DOTA and DOTA-peptide with 44m/44Sc and experimental evidence of the feasibility of an in vivo PET generator. Nucl. Med. Biol. 2014, 41, 36–43. [Google Scholar] [CrossRef]
  136. Domnanich, K.A.; Müller, C.; Farkas, R.; Schmid, R.M.; Ponsard, B.; Schibli, R.; Türler, A.; van der Meulen, N.P. Sc for labeling of DOTA- and NODAGA- functionalized peptides: Preclinical in vitro and in vivo investigations. EJNMMI Radiopharm. Chem. 2016, 1–19. [Google Scholar]
  137. Majkowska-Pilip, A.; Bilewicz, A. Macrocyclic complexes of scandium radionuclides as precursors for diagnostic and therapeutic radiopharmaceuticals. J. Inorg. Biochem. 2011, 105, 313–320. [Google Scholar] [CrossRef]
  138. Bouyer, F.; Sanson, N.; Destarac, M.; Gérardin, C. Hydrophilic block copolymer-directed growth of lanthanum hydroxide nanoparticles. New, J. Chem. 2006, 30, 399–408. [Google Scholar] [CrossRef]
  139. Wood, S.A.; Samson, I.M. The aqueous geochemistry of gallium, germanium, indium and scandium. Ore Geol. Rev. 2006, 28, 57–102. [Google Scholar] [CrossRef]
  140. Pyrzyńska, K.; Kilian, K.; Pęgier, M. Separation and purification of scandium: From industry to medicine. Sep. Purif. Rev. 2019, 48, 65–77. [Google Scholar] [CrossRef]
  141. Velikyan, I. 68Ga-based radiopharmaceuticals: Production and application relationship. Molecules 2015, 20, 12913–12943. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Applied Radiology. FDA Approves First Gallium 68Ga-DOTATATE Radiopharmaceutical Tracer for PET Imaging of Neuroendocrine Tumors. Available online: https://appliedradiology.com/articles/fda-approves-first-gallium-68ga-dotatate-radiopharmaceutical-tracer-for-pet-imaging-of-neuroendocrine-tumors (accessed on 30 January 2020).
  143. Aslani, A.; Snowdon, G.M.; Bailey, D.L.; Schembri, G.P.; Bailey, E.A.; Roach, P.J. Gallium-68 DOTATATE Production with Automated PET Radiopharmaceutical Synthesis System: A Three Year Experience. Asia Ocean. J. Nucl. Med. Biol. 2014, 2, 75–86. [Google Scholar]
  144. Decristoforo, C.; Knopp, R.; von Guggenberg, E.; Rupprich, M.; Dreger, T.; Hess, A.; Virgolini, I.; Haubner, R. A fully automated synthesis for the preparation of 68Ga-labelled peptides. Nucl. Med. Commun. 2007, 28, 7–12. [Google Scholar] [CrossRef] [PubMed]
  145. Ocak, M.; Antretter, M.; Knopp, R.; Kunkel, F.; Petrik, M.; Bergisadi, N.; Decristoforo, C. Full automation of 68Ga labelling of DOTA-peptides including cation exchange prepurification. Appl. Radiat. Isot. 2010, 68, 297–302. [Google Scholar] [CrossRef] [PubMed]
  146. Bailey, D.L.; Hofman, M.S.; Forwood, N.J.; O’Keefe, G.J.; Scott, A.M.; Van Wyngaardt, W.M.; Howe, B.; Kovacev, O.; Francis, R.J. Accuracy of dose calibrators for 68Ga PET imaging: Unexpected findings in a multicenter clinical pretrial assessment. J. Nucl. Med. 2018, 59, 636–638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. European Pharmacopoeia Organisation. Gallium (68Ga) Chloride Solution for Radiolabelling, European Pharmacopoeie 10.0; EDQM Council of Europe: Strasbourg, France, 2016; pp. 1206–1208. [Google Scholar]
  148. Radiopharmacy: An Update; A Technologist’s guide; EANM: Vienna, Austria, 2019; p. 149.
  149. Singh, A.; van der Meulen, N.P.; Müller, C.; Klette, I.; Kulkarni, H.R.; Tu, A.; Schibli, R.; Baum, R.P. First-in-Human PET/CT Imaging of Metastatic Neuroendocrine Neoplasms with Cyclotron-Produced 44Sc-DOTATOC: A Proof-of-Concept Study. Cancer Biother. Radiopharm. 2017, 32, 124–132. [Google Scholar] [CrossRef] [PubMed]
  150. Dvorakova, Z. Production and Chemical Processing of 177 Lu for Nuclear Medicine at the Munich Research Reactor FRM-II. Available online: https://www.researchgate.net/publication/36420376_Production_and_chemical_processing_of_Lu-177_for_nuclear_medicine_at_the_Munich_research_reactor_FRM-II (accessed on 30 January 2020).
  151. Lehenberger, S.; Barkhausen, C.; Cohrs, S.; Fischer, E.; Grünberg, J.; Hohn, A.; Köster, U.; Schibli, R.; Türler, A.; Zhernosekov, K. The low-energy β − and electron emitter 161Tb as an alternative to Lu for targeted radionuclide therapy. Nucl. Med. Biol. 2011, 38, 917–924. [Google Scholar] [CrossRef]
  152. Ferreira, K.M.; Collins, S.M.; Fenwick, A.J. Half-life measurement of the medical radioisotope 177Lu produced from the 176Yb(n, γ) reaction. EPJ Web Conf. 2017, 146, 1–5. [Google Scholar] [CrossRef] [Green Version]
  153. Duran, M.T.; Juget, F.; Nedjadi, Y.; Bochud, F.; Grundler, P.V.; Gracheva, G.; Müller, C. Determination of 161Tb half-life. J. Appl. Isot. 2020, in press. [Google Scholar] [CrossRef]
  154. Regulations for the Safe Transport of Radioactive Materials. IAEA Saf. Stand. 2012, SSR-6, 1–166.
  155. Nash, K.L.; Jensen, M.P. Analytical separations of the lanthanides: Basic chemistry and methods. Handb. Phys. Chem. Rare Earths 2000, 28, 311–371. [Google Scholar]
  156. Horwitz, E.P.; Mcalister, D.R.; Bond, A.H.; Barrans, R.E.; Williamson, J.M. A process for the separation of 177Lu from neutron irradiated 176Yb targets. Appl. Radiat. Isot. 63 2005, 63, 23–36. [Google Scholar] [CrossRef] [PubMed]
  157. van So, L.; Morcos, N.; Zaw, M.; Pellegrini, P.; Greguric, I. Alternative chromatographic processes for no-carrier added 177Lu radioisotope separation Part I. Multi-column chromatographic process for clinically applicable. J. Radioanal. Nucl. Chem. 2008, 277, 663–673. [Google Scholar]
  158. van So, L.; Morcos, N. New SPE column packing material: Retention assessment method and its application for the radionuclide chromatographic separation. J. Radioanal. Nucl. Chem. 2008, 277, 651–661. [Google Scholar]
  159. van So, L.; Morcos, N.; Zaw, M.; Pellegrini, P.; Greguric, I.; Nevissi, A. Alternative chromatographic processes for no-carrier added 177Lu radioisotope separation Part II. The conventional column chromatographic separation combined with HPLC for high purity. J. Radioanal. Nucl. Chem. 2008, 277, 675–683. [Google Scholar]
  160. Park, U.J.; Jae, U.; Lee, J.; Hyuk, K.; Soo, S.; Hyun, K. Lu-177 preparation for radiotherapy application. Appl. Radiat. Isot. 2016, 115, 8–12. [Google Scholar] [CrossRef]
  161. Marx, S.; Harfensteller, M.; Zhernosekov, K.; Nikula, T. Method of Manufacturing Non-Carrier Added High-Purity 177Lu Compounds as Well as Non-Carrier Added 177Lu Compounds. Patent US 2014/02, 2014. 1–8. Available online: https://patentimages.storage.googleapis.com/c2/d5/a3/32b542f3379064/US20140294700A1.pdf (accessed on 30 January 2020).
  162. Gracheva, N.; Müller, C.; Talip, Z.; Heinitz, S.; Köster, U.; Zeevaart, J.R.; Vögele, A.; Schibli, R.; van der Meulen, N.P. Production and characterization of no- carrier-added 161Tb as an alternative to the therapy. EJNMMI Radiopharm. Chem. 2019, 4, 1–16. [Google Scholar] [CrossRef]
  163. McAlister, D.R.; Horwitz, P.E. Characterization of extraction of chromatographic materials containing bis(2-ethyl-1-hexyl)phosphoric acid, 2-Ethyl-1-Hexyl (2-Ethyl-1-Hexyl) phosphonic acid, and bis(2,4,4-Trimethyl-1-Pentyl)phosphinic acid. Solvent Extr. Ion. Exch. 2007, 25, 757–769. [Google Scholar] [CrossRef]
  164. Horwitz, E.P.; McAlister, D.R.; Dietz, M.L. Extraction chromatography versus solvent extraction: How similar are they? Sep. Sci. Technol. 2006, 41, 2163–2182. [Google Scholar] [CrossRef]
  165. Starý, J. Separation of transplutonium elements. Talanta 1966, 13, 421–437. [Google Scholar] [CrossRef]
  166. Nayak, D.; Lahiri, S. Application of radioisotopes in the field of nuclear medicine. J. Radioanal. Nucl. Chem. 1999, 242, 423–432. [Google Scholar] [CrossRef]
  167. Cathy, C.; Jeffrey, S.; Ehrhardt, G.; Tyler, T.; Jurisson, S.; Deutsch, E. Current and Potential Therapeutic Uses of Lanthanide Radioisotope. Cancer Biother. Radiopharm. 2000, 15, 531–545. [Google Scholar]
  168. Rösch, F. Radiolanthanides in endoradiotherapy: An overview. Radiochim. Acta 2007, 95, 303–311. [Google Scholar] [CrossRef]
  169. Teo, R.D.; Termini, J.; Gray, H.B. Lanthanides: Applications in Cancer Diagnosis and Therapy Miniperspective. J. Med. Chem. 2016, 59, 6012–6024. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. van de Voorde, M.; van Hecke, K.; Cardinaels, T.; Binnemans, K. Radiochemical processing of nuclear-reactor-produced radiolanthanides for medical applications. Coord. Chem. Rev. 2019, 382, 103–125. [Google Scholar] [CrossRef]
  171. Iori, M.; Capponi, P.C.; Rubagotti, S.; Esposizione, L.R.; Seemann, J.; Pitzschler, R.; Dreger, T.; Formisano, D.; Grassi, E.; Fioroni, F.; et al. Labelling of 90Y- and 177Lu-DOTA-Bioconjugates for targeted radionuclide Therapy: A comparison among manual, semiautomated, and fully automated synthesis. Contrast Media Mol. Imaging 2017, 2017, 1–12. [Google Scholar] [CrossRef] [Green Version]
  172. Aslani, A.; Snowdon, G.M.; Bailey, D.L.; Schembri, G.P.; Bailey, E.A.; Pavlakis, N.; Roach, P.J. Lutetium-177 DOTATATE Production with an Automated Radiopharmaceutical Synthesis System. Asia Ocean. J. Nucl. Med. Biol. 2015, 3, 107–10715. [Google Scholar]
  173. European Pharmacopoeia Organisation. European Pharmacopoeia, Lutetium (177Lu) Solution for Radiolabelling. 2016, pp. 1218–1219. Available online: https://www.esrr.info/wp-content/uploads/2016/04/Kroon.pdf (accessed on 30 January 2020).
Figure 1. Stepwise radionuclide production of medical radionuclides (T1/2: half-life, σ: cross-section, Iγ and Iβ branching ratios).
Figure 1. Stepwise radionuclide production of medical radionuclides (T1/2: half-life, σ: cross-section, Iγ and Iβ branching ratios).
Molecules 25 00966 g001
Table 1. Decay characteristics and applications of radiometals currently used in clinical practice (decay data are taken from [22] and only the main γ-emission lines are shown).
Table 1. Decay characteristics and applications of radiometals currently used in clinical practice (decay data are taken from [22] and only the main γ-emission lines are shown).
RadionuclideHalf-LifeMain Decay ModeEα [MeV]av [MeV]Eγ [KeV]Iγ [%]Application
64Cu12.70 hβ+ 0.278511
1345.8
(annihil.)
0.48
PET
67Ga9.26 dγ 93.3
184.6
300.2
393.5
208.9
38.8
21.4
16.6
4.6
2.5
SPECT
68Ga67.71 minβ+ 0.830511
1077.3
(annihil.)
3.22
PET
111In2.80 dγ 245.3
171.3
94.1
90.7
SPECT
177Lu6.65 dβ 0.134208.4
112.9
10.4
6.2
β-Therapy
153Sm46.50 hβ 0.224103.2
69.7
29.3
4.7
Bone metastases
89Sr50.53 dβ 0.587 Bone
metastases
223Ra11.43 dα5.716
5.606
5.539
5.747
269.5
154.2
323.9
144.2
13.9
5.7
3.9
3.3
α-Therapy
186Re3.72 dβ 0.347137.29.5Bone metastases
99mTc6.01 hγ 140.589SPECT
201Tl72.91 hγ 167.4
135.3
10.0
2.6
SPECT
90Y64.10 hβ 0.934 β-Therapy
89Zr78.41 hβ+ 0.396511
909.1
(annihil.)
99
PET
Table 2. Comparison of 44Sc and 68Ga decay characteristics and production routes.
Table 2. Comparison of 44Sc and 68Ga decay characteristics and production routes.
RadionuclideHalf-lifeDecay modeEγ (kev)Main Production RouteRadionuclidic impurities
68Ga EC (11%) 68Ge →68Ga generator68Ge
67Ga, 66Ga,
 
67.71 min β+ (88%) 1077 (3%)68Zn(p,n)68Ga
+av: 830 keV
44Sc EC (5%), 44Ti → 44Sc generator48Sc, 44mSc
4.04 hβ+ (95%) 1157 (99.9%) natCa(p, xn)44Sc44mSc, 47Sc, 48Sc
+av: 632 keV 44Ca(p, n)44Sc44mSc
Table 3. Target material used for 44Sc and 68Ga production, respectively.
Table 3. Target material used for 44Sc and 68Ga production, respectively.
68GaReferences44ScReferences
Liquid TargetLiquid target
68ZnCl2[93]Ca(NO3)2[94]
68Zn(NO3)2[42,95,96,97]
Solid Target Solid target
68Zn foil[98]44CaCO3[99,100]
EP 68Zn[101,102,103]44CaO[104]
EP: Electroplated
Table 4. Specification for generator-produced 68Ga [147].
Table 4. Specification for generator-produced 68Ga [147].
Final product68GaCl3
AppearanceClear, colorless solution
pH<2
Radiolabeling Yield>99%
TLC/HPLC
Radionuclide Identity
(γ-Spectrometry Approx. Half Life)
511 keV
1077 keV
Radionuclidic purity
(γ-spectrometry)
>99.9%
Radiochemical Purity
(TLC)
>95%
68Ge Breakthrough<0.001%
Bacterial Endotoxins<175IU/V
Iron<10ug/GBq, each
Zinc
Table 5. Radionuclidic specifications based on the European Pharmacopoeia for generator and cyclotron-produced 68Ga [table adapted from [148].
Table 5. Radionuclidic specifications based on the European Pharmacopoeia for generator and cyclotron-produced 68Ga [table adapted from [148].
Final product Generator-produced 68GaCyclotron-produced 68Ga
68Ga activityMinimum 99.9%Minimum 98.0%
68Ge activityMaximum 0.001%n.a.
66Ga and 67Ga n.a.Maximum 2.0%
Other radioimpuritiesn.a.Maximum 0.1%
n.a. not applicable
Table 6. Comparison of 177Lu and 161Tb decay characteristics and production routes (data are taken from [22]).
Table 6. Comparison of 177Lu and 161Tb decay characteristics and production routes (data are taken from [22]).
RadionuclideHalf-Life (d)Eβ-av (MeV)Eγ (kev)Auger/Conversion e-Production Route
177Lu6.64 [152]134208 (10.4%)Auger L6.3 (8.7%)176Lu(n,γ)177Lu
113 (6.4%)CE K47.6 (5.1%)176Yb(n,γ)177Yb →177Lu
321 (0.219%)CE L101.7 (6.8%)
161Tb6.96 [153]15426 (23.2%)CE K3.4 (17.5%)160Gd(n,γ)161Gd →161Tb
29 (0.0365%)CE L16.6 (41%)
44 (0.060%)Auger L5.2 (87.9%)
Table 7. Comparison of neutron activation products of 176Lu, 176Yb, and 160Gd.
Table 7. Comparison of neutron activation products of 176Lu, 176Yb, and 160Gd.
Target% Natural
Abundance
% Available
Enrichment
Cross Section
σ (Barn)
Activation
Product
Decay
Mode
T1/2Decay
Product
Specific Activity
(TBq/mg)
176Lu2O32.5984.6 2.8177mLuβ, γ, IT160.4 d177Hf (78.6%)
177Lu (21.4%)→
3.9 TBq/mg
2090177Luβ, γ6.65 d177Hf
176Yb2O312.76>99.6 2.85177Ybβ, γ1.9 h177Lu740-1110 GBq/mg
160Gd2O321.8698.2 1.5161Gdβ, γ3.66 m161Tbto be optimized
Table 8. Comparison of the specification for 177LuCl3 and 161TbCl3 [162,173].
Table 8. Comparison of the specification for 177LuCl3 and 161TbCl3 [162,173].
Test177LuCl3 (EndolucinBeta)161TbCl3
Specific activity
(Dose calibrator)
36–44 GBq/mL at ART11–21 GBq/mL
AppearanceClear, colorlessClear, colorless
pH
(pH indicator strip)
1–21–2
Radiolabeling yield>99.0%>99.0%
TLC/HPLC
(Based on radiolabeling with
177Lu or 161Tb of DOTA-derivate, molar ratio 1:4)
Identity
(γ-spectrometry)
113 keV
208 keV
48.9 keV
74.6 keV
Radionuclidic purity
(γ-spectrometry)
175Yb ≤ 0.1%160Tb ≤ 0.007%
Radiochemical purity
(radio-TLC)
>99.0%>99.0%
Bacterial endotoxins≤175 IU/V
ART: Activity reference time.

Share and Cite

MDPI and ACS Style

Talip, Z.; Favaretto, C.; Geistlich, S.; Meulen, N.P.v.d. A Step-by-Step Guide for the Novel Radiometal Production for Medical Applications: Case Studies with 68Ga, 44Sc, 177Lu and 161Tb. Molecules 2020, 25, 966. https://doi.org/10.3390/molecules25040966

AMA Style

Talip Z, Favaretto C, Geistlich S, Meulen NPvd. A Step-by-Step Guide for the Novel Radiometal Production for Medical Applications: Case Studies with 68Ga, 44Sc, 177Lu and 161Tb. Molecules. 2020; 25(4):966. https://doi.org/10.3390/molecules25040966

Chicago/Turabian Style

Talip, Zeynep, Chiara Favaretto, Susanne Geistlich, and Nicholas P. van der Meulen. 2020. "A Step-by-Step Guide for the Novel Radiometal Production for Medical Applications: Case Studies with 68Ga, 44Sc, 177Lu and 161Tb" Molecules 25, no. 4: 966. https://doi.org/10.3390/molecules25040966

Article Metrics

Back to TopTop