Skip to main content
  • Original research
  • Open access
  • Published:

A GATE simulation study for dosimetry in cancer cell and micrometastasis from the 225Ac decay chain

Abstract

Background

Radiopharmaceutical therapy (RPT) with alpha-emitting radionuclides has shown great promise in treating metastatic cancers. The successive emission of four alpha particles in the 225Ac decay chain leads to highly targeted and effective cancer cell death. Quantifying cellular dosimetry for 225Ac RPT is essential for predicting cell survival and therapeutic success. However, the leading assumption that all 225Ac progeny remain localized at their target sites likely overestimates the absorbed dose to cancer cells. To address limitations in existing semi-analytic approaches, this work evaluates S-values for 225Ac’s progeny radionuclides with GATE Monte Carlo simulations.

Methods

The cellular geometries considered were an individual cell (10 µm diameter with a nucleus of 8 µm diameter) and a cluster of cells (micrometastasis) with radionuclides localized in four subcellular regions: cell membrane, cytoplasm, nucleus, or whole cell. The absorbed dose to the cell nucleus was scored, and self- and cross-dose S-values were derived. We also evaluated the total absorbed dose with various degrees of radiopharmaceutical internalization and retention of the progeny radionuclides 221Fr (t1/2 = 4.80 m) and 213Bi (t1/2 = 45.6 m).

Results

For the cumulative 225Ac decay chain, our self- and cross-dose nuclear S-values were both in good agreement with S-values published by MIRDcell, with per cent differences ranging from − 2.7 to − 8.7% for the various radionuclide source locations. Source location had greater effects on self-dose S-values than the intercellular cross-dose S-values. Cumulative 225Ac decay chain self-dose S-values increased from 0.167 to 0.364 GyBq−1 s−1 with radionuclide internalization from the cell surface into the cell. When progeny migration from the target site was modelled, the cumulative self-dose S-values to the cell nucleus decreased by up to 71% and 21% for 221Fr and 213Bi retention, respectively.

Conclusions

Our GATE Monte Carlo simulations resulted in cellular S-values in agreement with existing MIRD S-values for the alpha-emitting radionuclides in the 225Ac decay chain. To obtain accurate absorbed dose estimates in 225Ac studies, accurate understanding of daughter migration is critical for optimized injected activities. Future work will investigate other novel preclinical alpha-emitting radionuclides to evaluate therapeutic potency and explore realistic cellular geometries corresponding to targeted cancer cell lines.

Background

Targeted radiopharmaceutical therapy (RPT) involves using antigen-targeted molecules that are conjugated with alpha, beta, or Auger-electron emitting radionuclides, injected to accumulate, irradiate, and selectively kill cancer cells while sparing surrounding non-target tissue [1]. The therapeutic application of alpha emitters is of interest due to their short range and high energy deposition that results in effective cell death of the targeted cells and minimal damage to neighbouring normal healthy tissue [1]. Therapies with alpha particles have two distinct advantages over beta- or Auger-RPTs. First, the short range of alpha radiation in human tissues corresponds to only a few cell diameters (< 0.1 mm) where DNA damage can occur [2]. Second, the high energy (~ 5 MeV) and high linear energy transfer (LET) (~ 100 keV/µm) of alpha emissions leads to highly effective cell killing via DNA double strand breaks [2]. Because alpha particles deliver highly cytotoxic radiation with a limited range, they are ideal for the treatment of smaller tumour burdens, micrometastatic disease, and disseminated disease while avoiding damage to non-targeted cells [3].

Actinium-225 (225Ac) is a promising candidate isotope for RPT due to it successive cascade of high energy emissions with a total alpha energy of 27.6 MeV throughout its decay series and its relatively long-lived half-life of 9.92 days [4]. 225Ac has also been validated as a therapeutic radionuclide in several preclinical human studies [5]. This isotope has been targeted against many cancers including prostate, leukaemia, glioma, neuroendocrine, and melanoma, with remarkable outcomes in tumour control [6,7,8,9,10,11,12,13]. 225Ac decays to stable 209Bi through six short-lived radionuclide daughters (see Fig. 1). The decay cascade of 225Ac yields four alpha particles, two beta particles, and two gamma emissions. Specifically, 225Ac (t1/2 = 9.92 d; 5.8 MeV α-particle) decays to 221Fr (t1/2 = 4.80 m; 6.3 MeV α-particle), 217At (t1/2 = 32.6 ms; 7.1 MeV α-particle), 213Bi (t1/2 = 45.6 m; 435 keV mean energy β-particle with 97.8% branching ratio or 5.9 MeV α-particle with 2.2% branching ratio), 213Po (t1/2 = 3.72 μs; 8.4 MeV α-particle), 209Tl (t1/2 = 2.16 m; 660 keV mean energy β-particle), 209Pb (t1/2 = 3.23 h; 198 keV mean energy β-particle) and 209Bi (stable). The four alpha-particles have energies ranging from 5.8 to 8.4 MeV with associated tissue ranges of 47 to 85 µm [14]. Additionally, the two β-particles from the decay chain have therapeutic effects at further physical distances than the alpha emissions. The gamma emissions throughout the 225Ac decay chain can be used to assess biodistribution through imaging from the decay of 221Fr (218 keV, 11.6% branching ratio) and 213Bi (440 keV, 26.1% branching ratio) [15, 16].

Fig. 1
figure 1

225Ac decay scheme to stable 209Bi

In the context of RPT with an alpha emitter, internal radiation dosimetry is necessary for several reasons: first, to understand the radiobiological response; second, to compare and evaluate the efficacy of various preclinical therapeutic radionuclides; and third, to optimize the administered activity to achieve a personalized treatment. The standard method for calculating radiation dose from administered activity has been formalized by the Medical Internal Radiation Dosimetry (MIRD) schema which is applicable at organ, sub-organ, voxel, multicellular, and cellular levels [17]. The S-value defines the mean absorbed dose to a target region per radionuclide decay in a source region [18]. The absorbed dose is thus a product of the cumulative radioactivity over a time of interest and the S-value. When targeting individual cells and small tumour metastases, the dose to individual cells becomes important as the range of the emitted alpha particles is comparable in scale to the diameter of a cell or a small cluster of cells. Comprehensive databases of cellular S-values for many radionuclides of interest with spherical cells of various sizes and radionuclide distributions have been published by MIRD [19].

The calculations of cellular S-values by MIRD are based on convolution integrals, a semi-analytic method which applies the continuous slowing down approximation (CSDA). To address the limitations of the CSDA methodology, Monte Carlo track-structure (MCTS) methods have been studied to evaluate a number of radionuclides at a cellular level via simulations of radiation transport [18, 20,21,22,23,24,25,26,27,28,29]. However, in most studies, the focus has been largely on beta-emitting radionuclides with simple decay schemes, while a limited number of dosimetry studies focused on alpha emitters or 225Ac, specifically [22, 30, 31].

With respect to 225Ac and its complex decay chain, it is important to quantify the absorbed dose contributions from each radionuclide in the decay chain to determine the effect that progeny migration from the targeted cell will have on the overall therapeutic dose. During alpha decay, the recoil typically imparts an energy of ~ 0.1 MeV to the daughter nuclide, which is orders of magnitude higher than a typical chemical binding energy. The result is a daughter radionuclide that is no longer chemically bound to the radiopharmaceutical complex. The transfer of daughter radiation dose to off-target sites depends on its half-life, diffusion properties, and affinity for organs at risk. The first progeny in the decay of 225Ac is 221Fr (t1/2 = 4.80 m), which is a potassium analogue and is excreted from the cell via Na+/K+ pumps, potentiating subsequent decays occurring outside the target cells. The longer lived 213Bi (t1/2 = 45.6 m) can migrate to the kidneys, which may cause renal toxicity [32, 33]. There are many efforts underway to mitigate daughter migration from the target site, including targeting rapid cellular internalization, localized administration, and encapsulations in nanoparticles [34]. However, for current 225Ac-based therapies, daughter migration is often the limiting factor on injected activity and, consequently, therapeutic dose [34,35,36].

Here, we apply a MCTS approach to alpha emitters and, more importantly, to the complex 225Ac decay chain to discern the effects of radiopharmaceutical internalization and daughter migration. In this in silico study, we aim at evaluating the absorbed dose for radionuclides in the 225Ac decay chain (225Ac, 221Fr, 217At, 213Bi, 213Po, 209Tl, and 209Pb) for an individual cell and a cellular cluster to represent a micrometastatic tumour. To account for different levels of internalization of a radiopharmaceutical, we characterize the intra- and intercellular absorbed dose depositions to the cell nucleus for various subcellular distributions. We analyse the self- and cross- dose contributions. The dependency of S-values on the degree of radiopharmaceutical internalization and daughter radionuclide retention is also evaluated. The data presented here can assist in dosimetry calculations for novel 225Ac radiopharmaceuticals as they are evaluated in preclinical settings.

Methods

Monte Carlo simulations in GATE

We computed absorbed doses with simulations of radiation tracks in matter using the Monte Carlo simulations software GATE (Geant4 Application for Tomographic Emission) version 9.0. GATE uses physics modelling based on Geant4 (GEometry ANd Tracking) version 10.6.1 [37]. This toolkit was selected because of its well validated features for internal dosimetry in nuclear medicine [38,39,40].

All simulations used the low energy extended electromagnetic physics list emDNAphysics based on G4EmDNAPhysics (default constructor) of Geant4-DNA. The physical models loaded include all relevant electromagnetic physics processes, the interactions of all generated particles, and associated models required for the simulations. It enables collision-by-collision simulation of electron tracks down to the excitation threshold of liquid water (7.4 eV). We used the Geant4-DNA physics list because of its suitability for microdosimetry applications [41, 42] and good agreement with data obtained using other Monte Carlo codes like CELLDOSE, MC4V, PENELOPE, MCNP, and EGSnrc [27].

Modelled cellular geometries

We considered two cellular geometries: an individual cell and a cluster of cells that represent a micrometastasis. The single cell model consists of two concentric spheres representing the cell and the cell nucleus. The cell has a 10 µm diameter while the nucleus has an 8 µm diameter (See Fig. 2a). This geometry is based on formalized MIRD models and previous studies, to allow for direct comparison to previously established S-values [18, 27, 28]. The micrometastasis model consists of a cluster of individual cells (see Fig. 2b). Previous studies have considered a simple cubic structure model; however, we considered a hexagonal lattice as this provides more biologically reasonable spacing [43, 44]. Following a 3D hexagonal close packed lattice, the cluster consisted of a central cell with 12 neighbouring cells that are equidistance from the central cell. The central and neighbouring cells are simulated with the same cellular geometry as the isolated cell described previously. The material used for all cellular components was G4_WATER which is included in the GATE material database with a density of 1 g/cm3. While this cellular geometry simplifies the true in vivo structure of cancer cells and micrometastasis, it facilitates direct comparison to existing values derived from a semi-analytic method to establish the validity of Geant4-DNA Monte Carlo simulations for more interesting realistic cellular geometries in future works.

Fig. 2
figure 2

(left) Single cell of 10 µm diameter with a nucleus of 8 µm diameter. (right) Cell cluster modelled in this work. Dark blue represents the cell membrane and light blue represents the cell nucleus

225Ac decay chain

We individually simulated the decays of each radionuclide in the 225Ac decay scheme (see Fig. 1) to quantify the absorbed dose contributions with respect to each nuclide decay. Each radionuclide of interest is uniformly distributed in one of the following regions of the cell: (i) only on the cell membrane, (ii) only in the cytoplasm, (iii) only in the cell nucleus, or (iv) throughout the whole cell. There is equal radionuclide distribution in all cells of the cluster. GATE’s Radioactive Decay Module (RDM) simulates radioactive decays with secondary particle emissions on a per decay level [45, 46]. RDM produces primary particles using data from the evaluated nuclear structure data file (ENSDF) including branching ratios, decay energies and transition probabilities, and energies for nuclear de-excitation. RDM simulates the decay of radioactive nuclei through α, β, β+, isomeric transition, and electron capture. RDM simulates the entire decay and its associated decay emissions. With the GATE particleFilter tool, we can filter the atomic number (Z) and mass number (A) of the daughter progeny to stop the decay chain from continuing. The KillActor tool removes the daughter progeny from the simulation, so that we can simulate the dose deposition from each radionuclide individually.

The radionuclide source was input with source definitions (225Ac, 221Fr, 218At, 213Bi, 213Po, 209Tl, and 209Pb) and source distributions, or source location, (cell membrane, cytoplasm, nucleus, and whole cell). In all simulations, the total number of primary particles simulated was defined as 100,000 for alpha-emitting radionuclides (225Ac, 221Fr, 218At, 213Po) and 2,000,000 for beta-emitting radionuclides (213Bi, 209Tl, and 209Pb). Each combination of radionuclide and source distribution was simulated with Mersenne Twister random number generator [47] with each combination split into 10 separate jobs to reduce total run time. The number of primaries for alpha versus beta emission was determined to achieve comparable counting statistics, low uncertainties in the absorbed dose, and attainable simulation run times.

Measuring the absorbed dose

The GATE DoseActor tool scored the absorbed dose to each voxel, and its associated statistical uncertainty. This information is stored in a 3D matrix, or image, which enables fast visualization of the dose distribution. In our simulations, we scored the absorbed dose only to the nucleus, as it is the primary target for radiation-induced cell death [48]. For the cluster simulations, we scored the absorbed dose to the nucleus of the central cell, denoted in red in Fig. 2b. Given the geometrical symmetry of the cluster, the dose absorbed in the central cell is equally contributed from all 12 nearest neighbouring cells. Our voxel resolution was set to a matrix size of 100 × 100 × 100, with an individual voxel with dimensions of 0.8 µm × 0.8 µm × 0.8 µm.

The output of each simulation was a 3D matrix file containing the dose to each voxel. The dose volumetric data were analysed with Python v3.8.8 to calculate the total dose and associated uncertainties from the generated output files. The dose output images from each run were averaged together, and the total absorbed dose to the nucleus \((D_{{{\text{nuc}}}} )\) was calculated by the mean value of dose in each voxel in the region.

$$D_{{{\text{nuc}}}} = \frac{{E_{{{\text{tot}}}} }}{{M_{{{\text{nuc}}}} }} = \frac{{\sum E_{{{\text{vox}}}} }}{{M_{{{\text{nuc}}}} }} = \frac{{\sum E_{{{\text{vox}}}} }}{{n m_{{{\text{vox}}}} }} = \frac{{\sum D_{{{\text{vox}}}} }}{n}$$

where \(E_{{{\text{tot}}}}\) is the total energy deposited in the nucleus, \(M_{{{\text{nuc}}}}\) is the total mass of the nucleus, \(E_{{{\text{vox}}}}\) is the energy deposited in an individual voxel, \(m_{{{\text{vox}}}}\) is the mass of an individual voxel, and \(n\) is the number of voxels in the nucleus region. GATE also scores the dose squared in each voxel which is used to calculate the uncertainty with several runs. The standard error in the mean dose in each voxel \(\left( {u\left[ {D_{{{\text{vox}}}} } \right]} \right)\) was calculated according to the following equation [49].

$$u\left[ {D_{{{\text{vox}}}} } \right] = \sqrt {\frac{1}{N - 1}\left( {\frac{{\sum D_{{{\text{vox}}}}^{2} }}{N} - \left( {\frac{{\sum D_{{{\text{vox}}}} }}{N}} \right)^{2} } \right)}$$

where \(N\) is the number of primaries simulated. The standard error in the dose to the nucleus (\(u\left[ {D_{{{\text{nuc}}}} } \right]\)) is calculated with the following equation.

$$u\left[ {D_{{{\text{nuc}}}} } \right] = \frac{{\sum u[D_{{{\text{vox}}}} ]}}{n}$$

The number of primaries simulated ensured that the relative uncertainty in total dose is < 0.5% for all simulations.

S-value calculations

According to the MIRD scheme, the mean absorbed dose in a target region (\(r_{{\text{T}}}\)), \(D\left( {r_{{\text{T}}} \leftarrow r_{{\text{S}}} } \right)\), due to the cumulated activity, \(A_{{\text{C}}}\), in a source region (\(r_{{\text{S}}}\)) is defined by the following formula:

$$D\left( {r_{{\text{T}}} \leftarrow r_{{\text{S}}} } \right) = A_{{\text{C}}} \times S\left( {r_{{\text{T}}} \leftarrow r_{{\text{S}}} } \right)$$

where \(S\left( {r_{{\text{T}}} \leftarrow r_{{\text{S}}} } \right)\), is known as the S-value and denotes the dose to the target region per decay in the source region [19]. The S-values were calculated in units of GyBq−1 s−1, by dividing the total dose by the number of primary particles simulated.

We calculated the self-dose S-values directly from the individual cell simulations and calculated the cross-dose S-values from the cluster simulations. When considering the absorbed dose to the centre cell in a micrometastasis, it receives self-dose, from radionuclides localized within the central cell, and cross-dose, from radionuclides localized within all neighbouring cells [50]. Thus, the self-dose S-value is needed to derive the cross-dose S-value from absorbed dose to the centre cell’s nucleus. The ‘total’ S-values for self- and cross-dose were also calculated by summing the dose contributions from all radionuclides in the 225Ac decay chain, with consideration for branching ratios (see Fig. 1). The cumulative S-values represent the dose absorbed from the decay of 225Ac with the assumption that all progeny nuclides stay localized to the initial decay site. Cellular S-values were compared to the values from MIRDcell v3.10 software (Rutgers New Jersey Medical School, Newark, NJ) [51].

Radionuclide retention and internalization

By individually simulating each radionuclide in the 225Ac decay chain, we can assess the effect of progeny retention. 221Fr (t1/2 = 4.8 m) and 213Bi (t1/2 = 45.6 m) are the progeny nuclides of most concern when considering in vivo redistribution since they contribute successive alpha emissions following the decay of 225Ac. Although 217At (t1/2 = 32.6 ms) and 213Po (t1/2 = 3.72 μs) are also alpha emitters, they have very short half-lives and can be approximated to deposit their dose in the same location as their parent nuclide. 209Pb (t1/2 = 3.23 h) has a longer half-life; however, since it is a beta emitter, it is less critical to the overall therapeutic dose and off-target toxicities. We calculated the total self-dose S-values as a function of 221Fr and 213Bi retention by varying the progeny dose contribution for each source distribution. We considered progeny retention as ranging from 100% (221Fr + progeny or 213Bi + progeny localized) to 0% (221Fr + progeny or 213Bi + progeny migrated) in 20% increments.

By simulating various subcellular localization of the radionuclide, we can assess the effect that radiopharmaceutical internalization has on the absorbed dose to the nucleus. We defined cellular internalization as the fraction of radionuclides fully internalized throughout the whole cell relative to radionuclides not internalized and bound to the cellular membrane. We calculated the total self-dose S-values as a function of cellular internalization, ranging from 0% (all radionuclides bound to cellular membrane) to 100% (all radionuclides internalized inside the cell) in 20% increments.

Results

Self-dose S-values

Table 1 shows the self-dose S-values of the 225Ac decay chain radionuclides, as well as a cumulative total for the entire 225Ac decay chain with appropriate consideration of branching ratios. Making intuitive sense, the S-values are lowest when the radionuclide is localized to the cell surface, and conversely, are highest when internalized within the nucleus (see Fig. 3). Throughout the 225Ac decay chain, alpha emissions contribute more absorbed dose than beta emissions (see Fig. 4). For the radionuclides decaying via alpha emission (225Ac, 221Fr, 217At, 213Po), there was good agreement between MIRDcell S-values, with percentage differences < − 12.1%. However, for the radionuclides which decayed via beta emissions (213Bi, 209Tl, 209Pb), the obtained S-values were significantly different from MIRDcell (up to − 70.9% for 209Tl). Overall, the total decay chain S-values obtained using Geant4-DNA physics were in good agreement with MIRDcell S-values with the per cent difference ranging from − 4.7 to − 6.9%.

Table 1 Intracellular self-dose S-values (GyBq−1 s−1) for nuclides in the 225Ac decay chain from different source distributions targeting the nucleus
Fig. 3
figure 3

Cell nucleus dose maps for progeny radionuclides in the 225Ac decay chain. Radionuclide localization is defined as either bound to the cell membrane, within the cytoplasm, within the nucleus, or throughout the whole cell and the absorbed dose to the cell nucleus is scored. (Note: Plots have been normalized to number of primaries simulated and are visualized with equivalent heat map scaling)

Fig. 4
figure 4

Self-dose S-value (GyBq−1 s−1) to the cell nucleus for 225Ac progeny radionuclides. The figure legend indicates the radionuclide’s localization as either bound to the cell membrane (blue), within the cytoplasm (yellow), within the cell nucleus (green), or throughout the whole cell (cyan). Error bars represent one standard error, all with relative uncertainty values < 0.5%. Note: the S-values have been represented on a logarithmic scale to display the absorbed dose contributions of both alpha- and beta-emitting radionuclides in the 225Ac decay chain

Cross-dose S-values

Table 2 shows the intercellular cross-dose S-values delivered by the 225Ac decay chain radionuclides, as well as a cumulative total for the entire 225Ac decay chain, to the nucleus of the nearest neighbouring cell. The cross-dose S-values are relatively independent of subcellular localization (see Fig. 5) and primarily depend on radiation properties, such as mean alpha or beta emission energy. Comparing the cross-dose S-values obtained with Geant4-DNA physics to MIRDcell S-values saw good agreement. The alpha-emitting radionuclides (225Ac, 221Fr, 217At, 213Po) are within < 10.9%. However, like the self-dose S-value results, the beta-emitting radionuclides (213Bi,209Tl, 209Pb) have larger deviations from the MIRDcell S-values, up to − 46.8% for 209Tl. Overall, the combined cross-dose S-values for the entire decay chain differ between 2.7 and 8.7% from their MIRDcell counterparts.

Table 2 Intercellular cross-dose S-values (GyBq−1 s−1) for radionuclides in the 225Ac decay chain from different source distributions targeting the cell nucleus of a neighbouring cell
Fig. 5
figure 5

Cross-dose S-value (GyBq−1 s−1) to the cell nucleus of a nearest neighbouring cell for 225Ac progeny radionuclides. The figure legend indicates the radionuclide’s localization as either bound to the cell membrane (blue), within the cytoplasm (yellow), within the cell nucleus (green), or throughout the whole cell (cyan). Error bars represent one standard error, all with relative uncertainty values < 0.5%. Note: the S-values have been represented on a logarithmic scale to display the absorbed dose contributions of both alpha- and beta-emitting radionuclides in the 225Ac decay chain

Radionuclide retention and internalization

The effects of 221Fr and 213Bi retention on the total self-dose S-values to the cell nucleus are seen in Fig. 6.

Fig. 6
figure 6

Total self-dose S-value (GyBq−1 s−1) to the cell nucleus with various source distributions as a function of 221Fr (left) and 213Bi (right) retention

Figure 7 shows the effect of cellular internalization on the S-values for 225Ac’s progeny radionuclides from cell membrane localization to whole cell localization. The S-values all increase with increasing cellular internalization, by as much as 30% for the alpha emitter 213Po.

Fig. 7
figure 7

Total self-dose S-value (GyBq−1 s−1) to the cell nucleus with as a function of cellular internalization. Note: the S-values have been represented on a logarithmic scale to display the absorbed dose contributions of both alpha- and beta-emitting radionuclides in the 225Ac decay chain

Discussion

RPT with alpha emitters has shown remarkable promise as an effective cancer treatment. 225Ac’s therapeutic efficacy comes from the four alpha emissions throughout its decay chain. Characterizing the dose delivered within a cancer cell is critical to understanding its therapeutic outcome. The S-value metric is a universal parameter for assessing a radionuclide’s absorbed dose and therapeutic efficacy. In this study, we characterized the S-values in a single cell and micrometastatic environment for radionuclides in the 225Ac decay chain. The effects of progeny migration and radiopharmaceutical internalization were also assessed.

Validation of GATE simulations to MIRDcell

Our calculated S-values were compared to published values from MIRDcell to validate Geant4-DNA for cellular dosimetry estimates in alpha-emitting radionuclides. We found that for the alpha-emitting radionuclides both the self-dose and cross-dose S-values have good agreement with MIRDcell values; however, the S-values for the beta emitters showed larger differences from published values (see Tables 1, 2). These larger differences can be accounted for due to the scale of the cells and the range of the low energy beta emissions [53]. This effect has also been seen for electron dose deposition when the size of cell is comparable to the CSDA electron range in liquid water [27]. As the mean beta energy decreased from 660 keV (209Tl) to 435 keV (213Bi) to 198 keV (209Tl), our S-values show an underestimation, appropriate estimation, and overestimation, respectively, relative to the MIRDcell S-values. For the beta-emitting radionuclides 131I, 90Y, and 177Lu, large per cent differences between MIRDcell and Geant4-DNA S-values have been seen, with deviations ranging between − 79 and + 67% [54].

MIRDcell derives S-values with an analytic approach using the CSDA to calculate the energy deposition of beta and alpha emissions. However, simulating delta ray production and low energy transport becomes important to accurately estimate cellular dosimetry for low energy beta emissions, and the CSDA is less suitable in these situations where the physical geometry is close to the scale of the range of the beta emissions. There are limitations to the convolution integral method used in MIRD S-value calculation at the cellular scale. The CSDA approach neglects the finite range of secondary electrons (δ-rays) and energy loss straggling which are increasingly important on subcellular scales. However, when considering the 225Ac chain and its therapeutic applications, the alpha emissions deliver several magnitudes higher doses than the beta emissions (see Fig. 4). Given the strong agreement between the MIRDcell and Geant4-DNA S-values for the alpha-emitting isotopes, Geant4-DNA is validated for cellular dosimetry with alpha emitters.

In the MIRD formalism, the geometry of cells is limited to a spherical concentric design which is only an approximation of cancer cell geometries. Different cancer cell lines exhibit different geometries (spherical, ellipsoidal, irregular), which affects cellular S-values [20, 55]. Šefl et al. (2015) showed that for Auger-emitting radionuclides the S-value differences can be large between spherical and irregular cell geometries [27]. Further investigation is required to determine the magnitude of this effect for alpha-emitting radionuclides. Additionally, the size of the cancer cell and cell nucleus are critical to cellular S-values and are very dependent on cancer cell line [56]. Since the overall 225Ac decay chain has been well validated with our simplified cellular models, Geant4-DNA simulations would be well suited to further investigate microdosimetry in realistic, complex cellular geometries to evaluate preclinical 225Ac radiopharmaceuticals.

Dose estimates as a function of daughter retention

The primary concern surrounding 225Ac-based alpha therapy is the retention of daughter nuclides at the targeted site. While this is an important consideration for toxicity and limiting the injected dose, there is a collateral effect in overestimating absorbed doses to targeted cells. S-values to the cell nucleus decrease by up to 72% when there is no retention of 221Fr. This scenario is most physically realizable when a radiopharmaceutical is targeting a cell membrane protein and does not undergo internalization into the cell. Similarly, S-values decrease by up to 21% when there is no retention of 213Bi at the targeted cell. This is a realistic scenario for all source distributions, given that 213Bi has a longer half-life of 45 min. Maximizing the retention of progeny radionuclides is actively being researched with liposome carriers [57], polymeric vesicles [58], and nanoparticles [59].

These results highlight the importance of quantifying daughter retention and migration in vivo to ensure accurate absorbed dose measurements are delivered to cancer cells. Progeny migration can be evaluated and quantified in preclinical 225Ac radiopharmaceuticals with a variety of methods including alpha-cameras [36, 60], iQID cameras [61, 62], gamma spectroscopy [63], and alpha spectroscopy [64]. With quantified daughter migration, the S-values we have reported can be applied to determine the absorbed dose more accurately in preclinical 225Ac radiopharmaceutical.

Effect of cellular internalization of the radiopharmaceutical

We considered only four radionuclide distributions as they are standardized in S-values from MIRD. It is noted that cell membrane and cytoplasmic uptake of radioligands are the most physically realistic situations for targeted radiopharmaceutical distribution. However, as novel alpha emitter radiopharmaceuticals are developed, nuclear internalization may become a more common consideration. Overall, nuclear internalization of a 225Ac radiopharmaceutical can result in a twofold increase in the cumulative S-value; from 0.167 GyBq−1 s−1 (cell surface only) to 0.364 GyBq−1 s−1 (fully internalized). With nuclear internalization, the cumulative S-value increases threefold to 0.494 GyBq−1 s−1.

Considering that 225Ac has been described as an in vivo alpha generator for therapy, it is critical to quantify the internalization of 225Ac radiopharmaceuticals, and account for it while performing microdosimetry estimates. Specific peptides and antibodies are selected to be targeted by radiopharmaceuticals to maximize cellular internalization. It is expected that internalization will vary with each cell-line and targeting moiety. To determine the absorbed dose for a specific radiopharmaceutical, it is necessary to perform an in vitro internalization assay to determine the component of radiopharmaceutical that is membrane bound versus internalized into the cell [65]. With quantified internalization metrics, our S-value results can be applied to determine absorbed dose more accurately with preclinical 225Ac radiopharmaceuticals. Given the importance of retaining progeny radionuclides and maximizing absorbed dose to the cell nucleus, cellular internalization must also be a priority for developing alpha-emitting radiopharmaceuticals.

Conclusion

Monte Carlo simulations in GATE (with Geant4-DNA) are a valuable tool for estimating cellular and micrometastatic absorbed doses. While previous work has been heavily focused on beta emitters, this work illustrates GATE’s applications for microdosimetry with alpha-emitting radionuclides. As 225Ac radiopharmaceuticals are developed, these results can help to inform the most optimal cellular targets for maximizing absorbed dose to the nucleus of cancer cells. We have also shown the significance of progeny nuclide retention at the targeted site and cellular internalization of the radiopharmaceutical to optimize therapeutic efficacy. Future work will investigate other promising alpha emitters and more realistic complex cellular geometries.

Availability of data and materials

The datasets generated and analysed in the current study are available from the corresponding author on reasonable request.

Abbreviations

RPT:

Targeted radiopharmaceutical therapy

LET:

Linear energy transfer

MIRD:

Medical Internal Radiation Dosimetry

CSDA:

Continuous slowing down approximation

MCTS:

Monte Carlo track-structure

GATE:

Geant4 Application for Tomographic Emission

Geant4:

GEometry ANd Tracking

RDM:

Radioactive Decay Module

ENSDF:

Evaluated nuclear structure data file

References

  1. Gudkov SV, Shilyagina NY, Vodeneev VA, Zvyagin AV. Targeted radionuclide therapy of human tumors. Int J Mol Sci. 2016;17:33.

    Article  Google Scholar 

  2. Morgenstern A, Apostolidis C, Kratochwil C, Sathekge M, Krolicki L, Bruchertseifer F. An overview of targeted alpha therapy with 225actinium and 213bismuth. Curr Radiopharm. 2018;11:200–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Kim Y-S, Brechbiel MW. An overview of targeted alpha therapy. Tumor Biol. 2012;33:573–90.

    Article  CAS  Google Scholar 

  4. Pommé S, Marouli M, Suliman G, Dikmen H, Van Ammel R, Jobbágy V, et al. Measurement of the 225Ac half-life. Appl Radiat Isot. 2012;70:2608–14.

    Article  PubMed  Google Scholar 

  5. Morgenstern A, Apostolidis C, Bruchertseifer F. Supply and clinical application of actinium-225 and bismuth-213. Semin Nucl Med. 2020;50:119–23.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Jurcic JG, Rosenblat TL, McDevitt MR, Pandit-Taskar N, Carrasquillo JA, Chanel SM, et al. Phase I trial of the targeted alpha-particle nano-generator actinium-225 (225Ac-lintuzumab) (anti-CD33; HuM195) in acute myeloid leukemia (AML). J Clin Oncol. 2011;29:6516.

    Article  Google Scholar 

  7. Garg R, Allen KJH, Dawicki W, Geoghegan EM, Ludwig DL, Dadachova E. 225Ac-labeled CD33-targeting antibody reverses resistance to Bcl-2 inhibitor venetoclax in acute myeloid leukemia models. Cancer Med. 2021;10:1128–40.

    Article  CAS  PubMed  Google Scholar 

  8. Królicki L, Bruchertseifer F, Kunikowska J, Koziara H, Pawlak D, Kuliński R, et al. Dose escalation study of targeted alpha therapy with [225Ac]Ac-DOTA-substance P in recurrence glioblastoma—safety and efficacy. Eur J Nucl Med Mol Imaging. 2021;48:3595–605.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Kratochwil C, Apostolidis L, Rathke H, Apostolidis C, Bicu F, Bruchertseifer F, et al. Dosing 225Ac-DOTATOC in patients with somatostatin-receptor-positive solid tumors: 5-year follow-up of hematological and renal toxicity. Eur J Nucl Med Mol Imaging. 2021;49:54–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Kratochwil C, Bruchertseifer F, Giesel FL, Weis M, Verburg FA, Mottaghy F, et al. 225Ac-PSMA-617 for PSMA-targeted α-radiation therapy of metastatic castration-resistant prostate cancer. J Nucl Med. 2016;57:1941–4.

    Article  CAS  PubMed  Google Scholar 

  11. Kratochwil C, Bruchertseifer F, Rathke H, Bronzel M, Apostolidis C, Weichert W, et al. Targeted alpha therapy of mCRPC with 225Actinium-PSMA-617: dosimetry estimate and empirical dose finding. J Nucl Med. 2017;58:1624–31.

    Article  CAS  PubMed  Google Scholar 

  12. Kratochwil C, Bruchertseifer F, Rathke H, Hohenfellner M, Giesel FL, Haberkorn U, et al. Targeted α-therapy of metastatic castration-resistant prostate cancer with 225Ac-PSMA-617: swimmer-plot analysis suggests efficacy regarding duration of tumor control. J Nucl Med. 2018;59:795–802.

    Article  CAS  PubMed  Google Scholar 

  13. Yang H, Zhang C, Yuan Z, Rodriguez-Rodriguez C, Robertson A, Radchenko V, et al. Synthesis and evaluation of a macrocyclic actinium-225 chelator, quality control and in vivo evaluation of 225Ac-crown-αMSH peptide. Chem Eur J. 2020;26:11435–40.

    Article  CAS  PubMed  Google Scholar 

  14. Sgouros G, Roeske JC, McDevitt MR, Palm S, Allen BJ, Fisher DR, et al. MIRD pamphlet no. 22 (Abridged): radiobiology and dosimetry of α-particle emitters for targeted radionuclide therapy. J Nucl Med. 2010;51:311–28.

    Article  CAS  PubMed  Google Scholar 

  15. McDevitt MR, Finn RD, Sgouros G, Ma D, Scheinberg DA. An 225Ac/213Bi generator system for therapeutic clinical applications: construction and operation. Appl Radiat Isot. 1999;50:895–904.

    Article  CAS  PubMed  Google Scholar 

  16. Robertson AKH, Ramogida CF, Rodríguez-Rodríguez C, Blinder S, Kunz P, Sossi V, et al. Multi-isotope SPECT imaging of the 225 Ac decay chain: feasibility studies. Phys Med Biol. 2017;62:4406.

    Article  CAS  PubMed  Google Scholar 

  17. Bolch WE, Eckerman KF, Sgouros G, Thomas SR, Brill AB, Fisher DR, et al. MIRD pamphlet no. 21: a generalized schema for radiopharmaceutical dosimetry-standardization of nomenclature. J Nucl Med. 2009;50:477–84.

    Article  CAS  PubMed  Google Scholar 

  18. Goddu SM, Howell RW, Rao DV. Cellular dosimetry: absorbed fractions for monoenergetic electron and alpha particle sources and S-values for radionuclides uniformly distributed in different cell compartments. J Nucl Med. 1994;35:303–16.

    CAS  PubMed  Google Scholar 

  19. Bolch WE, Bouchet LG, Robertson JS, Wessels BW, Siegel JA, Howell RW, et al. MIRD pamphlet no. 17: the dosimetry of nonuniform activity distributions—radionuclide S values at the voxel level. J Nucl Med. 1999;40:11S-36S.

    CAS  PubMed  Google Scholar 

  20. Salim R, Taherparvar P. Cellular S values in spindle-shaped cells: a dosimetry study on more realistic cell geometries using Geant4-DNA Monte Carlo simulation toolkit. Ann Nucl Med. 2020;34:742–56.

    Article  PubMed  Google Scholar 

  21. Hocine N, Chipana R, Sarda L. Comparison of MCNPX and MIRDcell in assessing self-dose and cross-dose delivered to cell nuclei and the development of a realistic geometric model. Int J Radiat Biol. 2020;96:1008–16.

    Article  CAS  PubMed  Google Scholar 

  22. Pinto GM, Bonifacio DAB, De Sá LV, Lima LFC, Vieira IF, Lopes RT. A cell-based dosimetry model for radium-223 dichloride therapy using bone micro-CT images and GATE simulations. Phys Med Biol. 2020. https://doi.org/10.1088/1361-6560/ab6b42.

    Article  PubMed  Google Scholar 

  23. Bordes J, Incerti S, Mora-Ramirez E, Tranel J, Rossi C, Bezombes C, et al. Monte Carlo dosimetry of a realistic multicellular model of follicular lymphoma in a context of radioimmunotherapy. Med Phys. 2020;47:5222–34.

    Article  CAS  PubMed  Google Scholar 

  24. Carrasco-Hernández J, Ramos-Méndez J, Faddegon B, Jalilian AR, Moranchel M, Vila-Rodríguez MA. Monte Carlo track-structure for the radionuclide Copper-64: characterization of S-values, nanodosimetry and quantification of direct damage to DNA. Phys Med Biol. 2020;65:155005.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Salim R, Taherparvar P. Monte Carlo single-cell dosimetry using Geant4-DNA: the effects of cell nucleus displacement and rotation on cellular S values. Radiat Environ Biophys. 2019;58:353–71.

    Article  CAS  PubMed  Google Scholar 

  26. Alcocer-Ávila ME, Ferreira A, Quinto MA, Morgat C, Hindié E, Champion C. Radiation doses from 161Tb and 177Lu in single tumour cells and micrometastases. EJNMMI Phys. 2020;7:33.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Šefl M, Incerti S, Papamichael G, Emfietzoglou D. Calculation of cellular S-values using Geant4-DNA: the effect of cell geometry. Appl Radiat Isot. 2015;104:113–23.

    Article  PubMed  Google Scholar 

  28. Bousis C, Emfietzoglou D, Hadjidoukas P, Nikjoo H. Monte Carlo single-cell dosimetry of Auger-electron emitting radionuclides. Phys Med Biol. 2010;55:2555–72.

    Article  CAS  PubMed  Google Scholar 

  29. Barberet P, Vianna F, Karamitros M, Brun T, Gordillo N, Moretto P, et al. Monte-Carlo dosimetry on a realistic cell monolayer geometry exposed to alpha particles. Phys Med Biol. 2012;57:2189–207.

    Article  CAS  PubMed  Google Scholar 

  30. Lee D, Li M, Bednarz B, Schultz MK. Modeling cell and tumor-metastasis dosimetry with the particle and heavy ion transport code system (PHITS) software for targeted alpha-particle radionuclide therapy. Radiat Res. 2018;190:236–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Amato E, Leotta S, Italiano A, Baldari S. A Monte Carlo approach to small-scale dosimetry of solid tumour microvasculature for nuclear medicine therapies with 223Ra-, 131I-, 177Lu- and 111In-labelled radiopharmaceuticals. Phys Med. 2015;31:536–41.

    Article  PubMed  Google Scholar 

  32. Jaggi JS, Kappel BJ, McDevitt MR, Sgouros G, Flombaum CD, Cabassa C, et al. Efforts to control the errant products of a targeted in vivo generator. Cancer Res. 2005;65:4888–95.

    Article  CAS  PubMed  Google Scholar 

  33. Miederer M, McDevitt MR, Sgouros G, Kramer K, Cheung N-KV, Scheinberg DA. Pharmacokinetics, dosimetry, and toxicity of the targetable atomic generator, 225Ac-HuM195, in nonhuman primates. J Nucl Med. 2004;45:129–37.

    CAS  PubMed  Google Scholar 

  34. de Kruijff RM, Wolterbeek HT, Denkova AG. A critical review of alpha radionuclide therapy—how to deal with recoiling daughters? Pharmaceuticals. 2015;8:321–36.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Scheinberg DA, McDevitt MR. Actinium-225 in targeted alpha-particle therapeutic applications. Curr Radiopharm. 2011;4:306–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Cortez A, Josefsson A, McCarty G, Shtekler AE, Rao A, Austin Z, et al. Evaluation of [225Ac]Ac-DOTA-anti-VLA-4 for targeted alpha therapy of metastatic melanoma. Nucl Med Biol. 2020;88–89:62–72.

    Article  PubMed  Google Scholar 

  37. Sarrut D, Bardiès M, Boussion N, Freud N, Jan S, Létang JM, et al. A review of the use and potential of the GATE Monte Carlo simulation code for radiation therapy and dosimetry applications. Med Phys. 2014;41:064301.

    Article  PubMed  Google Scholar 

  38. Villoing D, Marcatili S, Garcia MP, Bardiès M. Internal dosimetry with the Monte Carlo code GATE: validation using the ICRP/ICRU female reference computational model. Phys Med Biol. 2017;62:1885–904.

    Article  CAS  PubMed  Google Scholar 

  39. Perrot Y, Degoul F, Auzeloux P, Bonnet M, Cachin F, Chezal JM, et al. Internal dosimetry through GATE simulations of preclinical radiotherapy using a melanin-targeting ligand. Phys Med Biol. 2014;59:2183–98.

    Article  CAS  PubMed  Google Scholar 

  40. Gupta A, Lee MS, Kim JH, Park S, Park HS, Kim SE, et al. Preclinical voxel-based dosimetry through GATE Monte Carlo simulation using PET/CT imaging of mice. Phys Med Biol. 2019;64:095007.

    Article  CAS  PubMed  Google Scholar 

  41. Champion C, Incerti S, Perrot Y, Delorme R, Bordage MC, Bardiès M, et al. Dose point kernels in liquid water: an intra-comparison between GEANT4-DNA and a variety of Monte Carlo codes. Appl Radiat Isot. 2014;83:137–41.

    Article  CAS  PubMed  Google Scholar 

  42. Francis Z, Incerti S, Capra R, Mascialino B, Montarou G, Stepan V, et al. Molecular scale track structure simulations in liquid water using the Geant4-DNA Monte-Carlo processes. Appl Radiat Isot. 2011;69:220–6.

    Article  CAS  PubMed  Google Scholar 

  43. Deroulers C, Aubert M, Badoual M, Grammaticos B. Modeling tumor cell migration: from microscopic to macroscopic models. Phys Rev E. 2009;79:031917.

    Article  Google Scholar 

  44. Iwasaki WM, Innan H. Simulation framework for generating intratumor heterogeneity patterns in a cancer cell population. PLoS ONE. 2017;12:e1005778.

    Article  Google Scholar 

  45. Hauf S, Kuster M, Batič M, Bell ZW, Hoffmann DHH, Lang PM, et al. Radioactive decays in Geant4. IEEE Trans Nucl Sci. 2013;60:2966–83.

    Article  Google Scholar 

  46. Hauf S, Kuster M, Batič M, Bell ZW, Hoffmann DHH, Lang PM, et al. Validation of Geant4-based radioactive decay simulation. IEEE Trans Nucl Sci. 2013;60:2984–97.

    Article  CAS  Google Scholar 

  47. Matsumoto M, Nishimura T. Mersenne twister: A 623-dimensionally equidistributed uniform pseudo-random number generator. ACM Trans Model Comput Simul. 1998;8:3–30.

    Article  Google Scholar 

  48. Franken NAP, ten Cate R, Krawczyk PM, Stap J, Haveman J, Aten J, et al. Comparison of RBE values of high-LET α-particles for the induction of DNA-DSBs, chromosome aberrations and cell reproductive death. Radiat Oncol. 2011;6:1–8.

    Article  Google Scholar 

  49. Chetty IJ, Rosu M, Kessler ML, Fraass BA, Ten Haken RK, Kong F-M, et al. Reporting and analyzing statistical uncertainties in Monte Carlo-based treatment planning. Int J Radiat Oncol. 2006;65:1249–59.

    Article  Google Scholar 

  50. Goddu SM, Rao DV, Howell RW. Multicellular dosimetry for micrometastases: dependence of self-dose versus cross-dose to cell nuclei on type and energy of radiation and subcellular distribution of radionuclides. J Nucl Med. 1994;35:521–30.

    CAS  PubMed  Google Scholar 

  51. Katugampola S, Wang J, Rosen A, Howell RW. MIRD pamphlet no. 27: MIRDcell V3, a revised software tool for multicellular dosimetry and bioeffect modeling. J Nucl Med. 2022;63:1441–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Vaziri B, Wu H, Dhawan AP, Du P, Howell RW, Bolch WE, et al. MIRD pamphlet no. 25: MIRDcell V2.0 software tool for dosimetric analysis of biologic response of multicellular populations. J Nucl Med. 2014;55:1557–64.

    Article  PubMed  Google Scholar 

  53. Tajik-Mansoury MA, Rajabi H, Mozdarani H. A comparison between track-structure, condensed-history Monte Carlo simulations and MIRD cellular S-values. Phys Med Biol. 2017;62:N90.

    Article  CAS  PubMed  Google Scholar 

  54. Tajik-Mansoury MA, Rajabi H, Mozdarani H. Cellular S-value of beta emitter radionuclide’s determined using Geant4 Monte Carlo toolbox, comparison to MIRD S-values. Iran J Nucl Med. 2016;24:37–45.

    CAS  Google Scholar 

  55. Tamborino G, De Saint-Hubert M, Struelens L, Seoane DC, Ruigrok EAM, Aerts A, et al. Cellular dosimetry of [177Lu]Lu-DOTA-[Tyr3]octreotate radionuclide therapy: the impact of modeling assumptions on the correlation with in vitro cytotoxicity. EJNMMI Phys. 2020;7:8.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Cai Z, Pignol J-P, Chan C, Reilly RM. Cellular dosimetry of 111In using Monte Carlo N-particle computer code: comparison with analytic methods and correlation with in vitro cytotoxicity. J Nucl Med. 2010;51:462.

    Article  PubMed  Google Scholar 

  57. Sofou S, Kappel BJ, Jaggi JS, McDevitt MR, Scheinberg DA, Sgouros G. Enhanced retention of the α-particle-emitting daughters of actinium-225 by liposome carriers. Bioconjug Chem. 2007;18:2061–7.

    Article  CAS  PubMed  Google Scholar 

  58. Wang G, de Kruijff RM, Rol A, Thijssen L, Mendes E, Morgenstern A, et al. Retention studies of recoiling daughter nuclides of 225Ac in polymer vesicles. Appl Radiat Isot. 2014;85:45–53.

    Article  CAS  PubMed  Google Scholar 

  59. Cędrowska E, Pruszynski M, Majkowska-Pilip A, Męczyńska-Wielgosz S, Bruchertseifer F, Morgenstern A, et al. Functionalized TiO2 nanoparticles labelled with 225Ac for targeted alpha radionuclide therapy. J Nanopart Res. 2018;20:83.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Bäck T, Jacobsson L. The α-camera: a quantitative digital autoradiography technique using a charge-coupled device for ex vivo high-resolution bioimaging of α-particles. J Nucl Med. 2010;51:1616–23.

    Article  PubMed  Google Scholar 

  61. Miller BW, Gregory SJ, Fuller ES, Barrett HH, Bradford Barber H, Furenlid LR. The iQID camera: an ionizing-radiation quantum imaging detector. Nucl Instrum Methods Phys Res Sect Accel Spectrom Detect Assoc Equip. 2014;767:146–52.

    Article  CAS  Google Scholar 

  62. Miller BW, Frost SHL, Frayo SL, Kenoyer AL, Santos E, Jones JC, et al. Quantitative single-particle digital autoradiography with α-particle emitters for targeted radionuclide therapy using the iQID camera. Med Phys. 2015;42:4094–105.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Tichacek CJ, Budzevich MM, Wadas TJ, Morse DL, Moros EG. A Monte Carlo method for determining the response relationship between two commonly used detectors to indirectly measure alpha particle radiation activity. Molecules. 2019;24:3397.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Engle JW, Weidner JW, Ballard BD, Fassbender ME, Hudston LA, Jackman KR, et al. Ac, La, and Ce radioimpurities in 225Ac produced in 40–200 MeV proton irradiations of thorium. Radiochim Acta. 2014;102:569–81.

    Article  CAS  Google Scholar 

  65. Qin Y, Imobersteg S, Blanc A, Frank S, Schibli R, Béhé MP, et al. Evaluation of actinium-225 labeled Minigastrin analogue [(225)Ac]Ac-DOTA-PP-F11N for targeted alpha particle therapy. Pharmaceutics. 2020;12:1088.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This research was enabled in part by support provided by the BC DRI Group and the Digital Research Alliance of Canada (alliancecan.ca).

Funding

This study was financially supported by the National Science and Engineering Research Council of Canada (NSERC) Discovery Grants RGPIN-2021-04093 (PS) and RGPIN-2021-02965 (CU). TRIUMF receives federal funding via a contribution agreement with the National Research Council of Canada (NRC).

Author information

Authors and Affiliations

Authors

Contributions

HK conducted simulations and data analysis. HK, CM, AR, PS, and CU contributed to the experimental design. HK drafted the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Helena Koniar.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Koniar, H., Miller, C., Rahmim, A. et al. A GATE simulation study for dosimetry in cancer cell and micrometastasis from the 225Ac decay chain. EJNMMI Phys 10, 46 (2023). https://doi.org/10.1186/s40658-023-00564-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40658-023-00564-5

Keywords