Skip to main content
Journal of Nuclear Medicine logoLink to Journal of Nuclear Medicine
. 2022 Jun;63(6):833–839. doi: 10.2967/jnumed.121.262671

Pretherapeutic Comparative Dosimetry of 177Lu-rhPSMA-7.3 and 177Lu-PSMA I&T in Patients with Metastatic Castration-Resistant Prostate Cancer

Benedikt Feuerecker 1,2,3,, Maythinee Chantadisai 1, Anne Allmann 1, Robert Tauber 4, Jakob Allmann 1, Lisa Steinhelfer 1, Isabel Rauscher 1, Alexander Wurzer 5, Hans-Jürgen Wester 5, Wolfgang A Weber 1,2, Calogero d’Alessandria 1,*, Matthias Eiber 1,2,*
PMCID: PMC9157737  PMID: 34531260

Visual Abstract

graphic file with name jnumed.121.262671absf1.jpg

Keywords: 177Lu-rhPSMA-7.3, 177Lu-PSMA I&T, dosimetry, mCRPC, prostate cancer, PSMA

Abstract

Radiohybrid prostate-specific membrane antigen (rhPSMA) ligands allow for labeling with 18F and radiometals for endoradiotherapy. rhPSMA-7.3 has been designated as a lead compound with promising preclinical data for 177Lu-rhPSMA-7.3, which has shown higher tumor uptake than 177Lu-PSMA I&T. In this retrospective analysis, we compared pretherapeutic clinical dosimetry data of both PSMA ligands. Methods: Six patients with metastatic castration-resistant prostate cancer underwent both 177Lu-rhPSMA-7.3 and 177Lu-PSMA I&T pretherapeutic dosimetry. Whole-body scintigraphy was performed at 1 h, 4 h, 24 h, 48 h, and 7 d after injection. Regions of interest covering the whole body, organs, bone marrow, and tumor lesions were drawn for each patient. Absorbed doses for individual patients and pretherapeutic applications were calculated using OLINDA/EXM. To facilitate the comparison of both ligands, we introduced the therapeutic index (TI), defined as the ratio of mean pretherapeutic doses to tumor lesions over relevant organs at risk. Results: Mean whole-body pretherapeutic effective doses for 177Lu-rhPSMA-7.3 and 177Lu-PSMA I&T were 0.12 ± 0.07 and 0.05 ± 0.03 Sv/GBq, respectively. Mean absorbed organ doses for 177Lu-rhPSMA-7.3 and 177Lu-PSMA I&T were, for example, 1.65 ± 0.28 and 0.73 ± 0.18 Gy/GBq for the kidneys, 0.19 ± 0.09 and 0.07 ± 0.03 Gy/GBq for the liver, 2.35 ± 0.78 and 0.80 ± 0.41 Gy/GBq for the parotid gland, and 0.67 ± 0.62 and 0.30 ± 0.27 Gy/GBq for the bone marrow, respectively. Tumor lesions received mean absorbed doses of 177Lu-rhPSMA-7.3 and 177Lu-PSMA I&T of 6.44 ± 6.44 and 2.64 ± 2.24 Gy/GBq, respectively. The mean TIs for the kidneys were 3.7 ± 2.2 and 3.6 ± 2.2 for 177Lu-rhPSMA-7.3 and 177Lu-PSMA I&T, respectively, and those for the bone marrow were 15.2 ± 10.2 and 15.1 ± 10.2 for 177Lu-rhPSMA-7.3 and 177Lu-PSMA I&T, respectively. Conclusion: Pretherapeutic clinical dosimetry confirmed preclinical results of mean absorbed doses for tumors that were 2–3 times higher for 177Lu-rhPSMA-7.3 than for 177Lu-PSMA I&T. Absorbed doses to normal organs also tended to be higher for 177Lu-rhPSMA-7.3, resulting overall in similar average TIs for both radiopharmaceuticals with considerable interpatient variability. 177Lu-rhPSMA-7.3 has promise for a therapeutic efficacy similar to that of 177Lu-PSMA I&T at smaller amounts of injected activity, simplifying radiation safety measurements (especially for large patient numbers or dose escalation regimens).


Treatment of metastatic castration-resistant prostate cancer (mCRPC) remains challenging. 177Lu-PSMA radioligand therapy (RLT) is an option with a variety of different prostate-specific membrane antigen (PSMA) ligands developed in recent years (1). Several prospective and retrospective studies proved that 177Lu-PSMA (using either PSMA-617 or PSMA I&T) had substantial antitumor effects (2,3). Most recently, the VISION trial showed longer median radiographic progression-free survival (8.7 vs. 3.4 mo) and overall survival (15.3 vs. 11.3 mo) for 177LuPSMA-617 versus the standard of care, respectively, in PSMA-positive mCRPC after the use of taxane and next-generation androgen receptor signaling inhibitor agents (4).

For the assessment of new radiopharmaceuticals, dosimetry is essential to link the potential range of injected activities with therapeutic responses and possible side effects. For example, PSMA ligands can exhibit intense tracer accumulation in some normal organs, such as the kidneys. Dosimetric results have been published for the theranostic DOTA-conjugated PSMA ligands 177Lu-PSMA-DKFZ-617 and 177Lu-PSMA I&T (5), including both pretherapeutic (6) and posttherapeutic (710) evaluations.

Recently, a class of radiohybrid PSMA (rhPSMA) ligands were developed. They are theranostic agents allowing both fluorination and labeling with radiometals (1114). Preclinical data have proposed that the single diastereoisomer 18F-rhPSMA-7.3 is the most promising clinical candidate (15,16). 18F-rhPSMA-7.3 is currently in 2 phase 3 trials for PET imaging of primary (NCT04186819) and recurrent (NCT04186845) prostate cancers. Most recently, promising preclinical data on 177Lu-rhPSMA-7.3 in comparison to 177Lu-PSMA I&T were published (16).

Here, we present a retrospective analysis exploring the potential of 177Lu-rhPSMA-7.3 in comparison to 177Lu-PSMA I&T for endoradiotherapy in mCRPC. We used pretherapeutic comparative dosimetry data for normal organs and tumor lesions.

MATERIALS AND METHODS

Patients and Rationale for Comparative Dosimetry

According to current German guidelines, mCRPC patients after chemotherapy and novel antiandrogen therapy can be considered for 177Lu-PSMA RLT after interdisciplinary tumor board discussion (17). All patients in the presented analysis had undergone chemotherapy and novel antiandrogen therapy before 177Lu-PSMA.

Patients were informed that there are no approved PSMA-targeted therapies but that preliminary preclinical and clinical data support the antitumor activity of 177Lu-PSMA I&T. Additionally, information about preclinical data showing higher uptake of 18F-rhPSMA-7.3 and higher absorbed doses of 177Lu-rhPSMA-7.3 in tumors (14,16), indicating higher radiation doses to tumor tissue of 177Lu-rhPSMA-7.3 than of 177Lu-PSMA I&T for clinical use, was provided.

Patients were offered pretherapeutic administration of both 177Lu-PSMA I&T and 177Lu-rhPSMA-7.3 to determine tumor and normal organ doses. Subsequent treatment was then performed with the agent that showed favorable tumor-to-normal organ dose ratios or with 177Lu-PSMA I&T if the differences in the tumor-to-normal organ dose ratios were similar. 177Lu-PSMA I&T and 177Lu-rhPSMA-7.3 were prepared in compliance with the German Medicinal Products Act, AMG §13 2b, and after informing the responsible regulatory body (Government of Oberbayern, Germany). The institutional review board of the Technical University of Munich approved the retrospective scientific analysis of the dosimetry data (115/18 S-KK).

Between April 2018 and November 2020, 6 patients agreed to undergo these dosimetric investigations. Patient characteristics are presented in Supplemental Table 1 (supplemental materials are available at http://jnm.snmjournals.org). The approach was based on the rationale of individual selection of the optimal ligand for a specific patient to offer the possibility of benefit from higher tumor uptake, as recent preclinical data indicated (16).

Definitions of Therapeutic Index (TI) and Relative TI (rTI)

To assess the potential antitumor effect in relation to organs at risk of 177Lu-PSMA I&T versus 177Lu-rhPSMA-7.3, a TI was calculated. It was defined as the mean radiation dose to tumor lesions divided by the radiation dose to relevant organs at risk. As the kidneys and bone marrow are the most relevant organs, we report the TI for the kidneys and the TI for the bone marrow (4). The respective rTI was defined as the ratio of the TI of 177Lu-rhPSMA-7.3 to the TI of 177Lu-PSMA I&T, with a value of greater than 1 indicating a distribution favoring 177Lu-rhPSMA-7.3.

Pretherapeutic Dosimetry, Image Analysis, and Dosimetric Calculations

The mean applied pretherapeutic activities were 1,066 ± 83 MBq (range, 1,000–1,243 MBq) for 177Lu-PSMA I&T and 1,012 ± 51 MBq (range, 917–1,083 MBq) for 177Lu-rhPSMA-7.3. Activity was injected over approximately 1 min and was followed by a saline flush. Specific activities were 47.5 GBq/0.59 μmol for 177Lu-PSMA I&T and 47.5 GBq/0.61 μmol for 177Lu-rhPSMA-7.3. The mean time period between application of both agents was 172 h (range, 166–190 h). Whole-body scintigraphy was performed at least 1 h, 4 h, 24 h, 48 h, and 7 d after administration.

Individual patient absorbed doses for the whole body, kidneys, liver, parotid, submandibular, and lacrimal glands, tumor lesions, and red bone marrow were estimated on the basis of the MIRD scheme, as recommended in the European Association of Nuclear Medicine Dosimetry Committee Guidelines. Absorbed organ and tumor doses for each cycle were calculated using OLINDA/EXM (1820). Details on the regions of interest (ROIs) for scintigraphy and the volume calculations for PET are given in the supplemental data (e.g., Supplemental Table 2).

Statistical Analysis

All continuous data reported are expressed as mean ± SD and range. A nonpaired t test followed by Welch correction was performed to compare means. Statistical analyses were conducted using GraphPad Prism (version 5.0; GraphPad Software).

RESULTS

Qualitative 177Lu-PSMA I&T and 177Lu-rhPSMA-7.3 Biodistributions on Pretherapeutic Scintigraphy

Physiologic uptake was seen in the lacrimal, parotid, and submandibular glands, kidneys, and small intestine; uptake was less pronounced in the liver and spleen. Uptake in excess of the background was also seen for multiple tumor lesions, with progressive accumulation up to 24–48 h after injection for 177Lu-PSMA I&T and 177Lu-rhPSMA-7.3 (Fig. 1). Delayed whole-body images (up to 7 d after therapy) exhibited long-term retention of 177Lu-PSMA I&T and 177Lu-rhPSMA-7.3 in the metastases, with nearly no residual uptake in normal organs.

FIGURE 1.

FIGURE 1.

Examples of ROIs in 1 patient for 177Lu-PSMA I&T (top) and 177Lu-rhPSMA-7.3 (bottom).

Pretherapeutic Dosimetry of Normal Organs

The mean whole-body pretherapeutic effective dose for 177Lu-rhPSMA-7.3 was 0.117 Gy (0.12 ± 0.07 Sv/GBq), and that for 177Lu-PSMA I&T was 0.054 Gy (0.05 ± 0.03 Sv/GBq). The mean absorbed organ doses for 177Lu-rhPSMA-7.3 and 177Lu-PSMA I&T were 1.65 ± 0.28 and 0.73 ± 0.18 Gy/GBq, respectively, for the kidneys; 0.19 ± 0.09 and 0.07 ± 0.03 Gy/GBq, respectively, for the liver; 2.35 ± 0.78 and 0.80 ± 0.41 Gy/GBq for the parotid glands, respectively; 2.10 ± 0.86 and 0.67 ± 0.31 Gy/GBq for the submandibular glands, respectively; and 5.29 ± 2.16 and 1.92 ± 0.80 Gy/GBq for the lacrimal glands, respectively (Supplemental Table 3). Figure 2 and Supplemental Figs. 1 and 2 display mean organ doses, individual organ doses, and individual percentage injected doses.

FIGURE 2.

FIGURE 2.

Mean organ doses (Gy/GBq) for kidneys, liver, parotid, lacrimal, and submandibular (Submand.) glands, and tumor lesions and total-body doses (Sv/GBq) determined with 177Lu-PSMA I&T (I&T) and 177Lu-rhPSMA-7.3 (rh) for all patients. Individual patient organ doses are shown in Supplemental Figs. 1–3. RM = red bone marrow. RM* = using an ROI for correction next to the lumbar spine (n = 4); RM** = using an ROI in the thigh (n = 6); #Sv/GBq.

Pretherapeutic Dosimetry of Bone Marrow

When ROIs were placed in the thigh regions, red bone marrow absorbed doses were 0.67 ± 0.62 Gy/GBq for 177Lu-rhPSMA-7.3 and 0.30 ± 0.27 Gy/GBq for 177Lu-PSMA I&T. Data for bone marrow dosimetry obtained with ROIs next to the lumbar spine for correction are presented in Supplemental Tables 3 and 4.

Pretherapeutic Dosimetry of Tumor Lesions

A total of 21 representative lesions were analyzed (14 bone and 7 lymph node metastases). Mean and individual sizes of individual tumor lesions are given in the supplemental materials and Supplemental Table 2.

The pretherapeutic mean absorbed doses of tumor lesions were 6.44 ± 6.44 Gy/GBq (range, 0.66–29.25 Gy/GBq) for 177Lu-rhPSMA-7.3 and 2.64 ± 2.24 Gy/GBq (range, 0.38–9.80 Gy/GBq) for 177Lu-PSMA I&T. The pretherapeutic mean absorbed doses for bone and lymph node metastases were 4.09 ± 2.57 and 11.14 ± 8.83 Gy/GBq, respectively, for 177Lu-rhPSMA-7.3 and 1.70 ± 1.13 and 4.51 ± 2.69 Gy/GBq, respectively, for 177Lu-rhPSMA-7.3 (Table 1). Figure 2 and Supplemental Figs. 1–3 display mean tumor doses, individual tumor doses, and individual percentage injected doses.

TABLE 1.

Effective Doses for Tumor and Ratios to Effective Doses for Kidney and Bone Marrow

177Lu-rhPSMA-7.3 (rh) 177Lu-PSMA I&T (I&T)
Patient Tumor no. Tumor site Tumor dose (mGy/MBq) TI for kidneys TI for bone marrow Tumor dose (mGy/MBq) TI for kidneys TI for bone marrow Ratio of tumor rh to tumor I&T rTI for kidneys rTI for bone marrow*
1 1 B 3.00 1.91 3.63 1.31 2.07 3.45 2.30 0.92 1.05
2 B 1.58 1.01 1.91 0.73 1.16 1.94 2.16 0.87 0.99
2 1 B 4.05 2.33 11.60 1.38 1.71 9.82 2.95 1.36 1.18
2 B 6.94 3.99 19.90 2.31 2.88 16.51 3.00 1.39 1.21
3 LN 29.25 16.81 83.81 9.80 12.20 70.00 2.98 1.38 1.20
4 B 2.81 1.62 8.05 0.88 1.09 6.27 3.20 1.48 1.28
5 B 1.53 0.88 4.37 0.52 0.65 3.72 2.94 1.35 1.18
3 1 LN 16.15 11.97 43.42 5.15 10.42 35.99 3.14 1.15 1.21
2 LN 14.00 10.37 37.63 5.93 12.01 41.49 2.36 0.86 0.91
3 B 1.56 1.16 4.20 0.73 1.47 5.07 2.15 0.79 0.83
4 1 B 2.21 1.24 8.24 0.92 0.97 8.45 2.40 1.28 0.98
2 LN 3.46 1.94 12.91 1.22 1.28 11.19 2.84 1.52 1.15
5 1 B 6.88 3.23 3.55 2.79 2.96 3.26 2.47 1.09 1.09
2 B 6.67 3.13 3.44 3.00 3.18 3.51 2.22 0.99 0.98
3 B 5.92 2.78 3.05 2.69 2.85 3.14 2.20 0.98 0.97
4 B 9.49 4.46 4.89 4.37 4.63 5.11 2.17 0.96 0.96
5 B 3.91 1.84 2.02 1.82 1.93 2.13 2.15 0.95 0.95
6 1 B 0.66 0.50 3.38 0.38 0.66 4.00 1.72 0.75 0.85
2 LN 8.05 6.05 41.28 4.66 8.06 48.54 1.73 0.75 0.85
3 LN 3.38 2.54 17.32 1.93 3.33 20.06 1.75 0.76 0.86
4 LN 3.71 2.79 19.03 2.89 5.00 30.10 1.28 0.56 0.63
Mean ± SD for all 6.44 ± 6.44 3.93 ± 4.07 16.08 ± 19.79 2.64 ± 2.24 3.83 ± 3.58 15.89 ± 18.28 2.39 1.05 1.01
Mean ± SD for B 4.09 ± 2.57§ 2.15 ± 1.17 5.87 ± 4.68 1.70 ± 1.13 2.02 ± 1.11 5.46 ± 3.76 2.43 1.08 1.03
Mean ± SD for LN 11.14 ± 8.83 7.50 ± 5.27 36.49 ± 22.49 4.51 ± 2.69 7.47 ± 4.03 36.77 ± 17.90 2.30 1.00 0.97
*

Results for red marrow dosimetry using ROI placed in thigh.

P = 0.02.

Not significant.

§P = 0.007.

B = bone; LN = lymph node.

Data for all patients are presented for individual tumor lesions and grouped by tumor lesion type.

TI and rTI

The mean TIs for the kidneys were 3.7 ± 2.2 for 177Lu-rhPSMA-7.3 and 3.6 ± 2.2 for 177Lu-PSMA I&T. Intraindividual comparisons of 177Lu-rhPSMA-7.3 and 177Lu-PSMA I&T revealed a higher TIs for the kidneys in 2 patients for 177Lu-rhPSMA-7.3 (patient 2: 5.1 vs. 3.7; patient 4: 1.6 vs. 1.1) and in 1 patient for 177Lu-PSMA I&T (patient 6: 2.9 vs. 4.3). In 3 patients, no clear differences were seen (patient 1: 1.5 vs. 1.6; patient 3: 7.8 vs. 7.9; patient 5: 3.1 vs. 3.1). Consequently, the rTI for the kidneys was greater than 1 in patients 2 and 4 and less than or equal to 1 in all other patients. The individual TIs and the rTIs are shown in Figures 3A and 3B, respectively.

FIGURE 3.

FIGURE 3.

TIs (A) and rTIs (B) for tumor-to-kidney ratio for each individual patient. Values of >1 indicate favorable biodistribution for 177Lu-rhPSMA-7.3 (rh) compared with 177Lu-PSMA I&T (I&T) and vice versa. Two patients had favorable distribution of 177Lu-rhPSMA-7.3, 1 patient had favorable distribution of 177Lu-PSMA I&T, and in 3 patients no clear preference was observed.

When ROIs in the thigh were used, the mean TIs for the bone marrow were 15.2 ± 10.2 for 177Lu-rhPSMA-7.3 and 15.1 ± 10.2 for 177Lu-PSMA I&T. Intraindividual comparisons of 177Lu-rhPSMA-7.3 and 177Lu-PSMA I&T revealed that 177Lu-rhPSMA-7.3 showed higher TIs for the bone marrow in 4 patients (patient 1: 2.8 vs. 2.7; patient 2: 25.5 vs. 21.3; patient 3: 28.4 vs. 27.5; patient 4: 10.6 vs. 9.8). In 1 patient, no clear difference was measured (patient 5: 3.4 vs. 3.4), and in another patient, 177Lu-PSMA I&T showed a higher TI for the bone marrow (patient 6: 20.3 vs. 25.7).

DISCUSSION

We presented data on pretherapeutic radiation dosimetry for normal organs and tumor lesions for 177Lu-rhPSMA-7.3 and 177Lu-PSMA I&T in 6 mCRPC patients. Quantitative analyses revealed, on average, an absorbed dose to tumor lesions of 177Lu-rhPSMA-7.3 that was 2.4 times higher than that of 177Lu-PSMA I&T. This finding is in line with recent preclinical data demonstrating a 2.6-fold difference (16). However, in our clinical investigation, absorbed doses to normal organs were also 2–3 times higher (e.g., 2.3 for the kidneys, 2.9 for the parotid glands, and 2.2 for the bone marrow). Notably, these relationships substantially differed at the patient level.

Because of controversies about the extrapolation of preclinical evaluations, pretherapeutic clinical dosimetry is important (21). Currently, dosimetry for PSMA ligands focuses on absorbed doses delivered to normal organs, primarily the kidneys but also the salivary glands as the most relevant organs at risk. Despite numerically high absorbed doses to the salivary and parotid glands, clinically relevant toxicity has only been anecdotally reported and has been mainly transient (22). Although red bone marrow dosing is essential, its methodology is prone to errors—for example, as a result of the frequent presence of extensive bone metastases. Nevertheless, bone marrow toxicity even in the presence of extensive osseous metastases is not a frequent side effect (4,23).

rhPSMA ligands belong to a new class of fully theranostic agents (16). They allow the use of radiochemical twins, such as 19F/177Lu-rhPSMA or 18F/natLu-rhPSMA, for potential pretherapeutic PET-based imaging and subsequent PSMA RLT (24). Recently published promising preclinical data demonstrated that the radiohybrid 19F/177Lu-rhPSMA-7.3 is a suitable candidate for clinical translation because of similar clearance kinetics and radiation doses but superior tumor uptake and retention compared with 177Lu-PSMA I&T (16). Using this approach, our aim was to investigate 177Lu-rhPSMA-7.3 by comparing it to 177Lu-PSMA I&T as the established agent for PSMA RLT, allowing us to maximize the absorbed radiation dose to tumor lesions and to minimize the absorbed radiation dose to relevant organs at risk.

When differences in radiation doses to normal organs are investigated, the kidneys are usually regarded as dose-limiting organs at risk. Pretherapeutic kidney doses in our 6 mCRPC patients were 2.3 times higher with 177Lu-PSMA-7.3 than with 177Lu-PSMA I&T. Regarding potential radiation damage, currently either 23 Gy with a 5% probability of late kidney damage within 5 y or 28 Gy with a 50% probability of late kidney damage within 5 y is used (25). For 177Lu-PSMA I&T and 177Lu-PSMA-617, severe kidney toxicity has been described as a side effect in only a few patients (26). However, care must be taken given the limited overall survival of late-stage mCRPC patients. Kabaskal et al. calculated a maximum activity of 32.9 GBq to achieve a 23-Gy kidney dose report for 177Lu-PSMA-617 using pretherapeutic dosimetry (10). For 177Lu-rhPSMA-7.3, our mean data would indicate the option to apply approximately 17 GBq on the basis of a mean of 1.65 Gy/GBq. However, this lower activity would achieve similar absorbed tumor doses. It remains to be decided which activity levels and timing of cycles will be pursued in any potential clinical development of 177Lu-rhPSMA-7.3. Nevertheless, we believe that the presented data might inform potential future study protocols.

Notably, given the results of the VISION trial, bone marrow toxicity is a rare but relevant side effect (4). In our retrospective study, the radiation delivery to the bone marrow was 2.2 times higher for 177Lu-rhPSMA-7.3. The relative TI of 1.2, integrating absorbed doses to both tumors and bone marrow, suggests only a slight improvement over 177Lu-PSMA I&T.

However, the calculation of red bone marrow doses is complex when based on scintigraphic images. For example, in 2 of our patients, radiation exposure of the bone marrow was probably overestimated because of the presence of tumor lesions in the ROI. Nevertheless, although important for absolute values, the ratio between 177Lu-rhPSMA-7.3 and 177Lu-PSMA I&T was probably less affected. Our results for bone marrow dosimetry showed mean absorbed doses of 0.67 ± 0.62 mGy/MBq for 177Lu-rhPSMA-7.3 and 0.30 ± 0.27 mGy/MBq for 177Lu-PSMA I&T. These doses resulted in a favorable tumor-to-bone marrow index in 3 patients. The absolute values were substantially higher than those reported in the literature for 177Lu-PSMA-617, but the difference can be mainly explained by the different methods (thigh vs. lumbar spine correction) (7,8).

It is important to emphasize that the absorbed dose limits for solid organs are based on conventionally fractionated external-beam therapy and cannot necessarily be directly applied to low-dose-rate radiation (27). Patients without risk factors for kidney disease might tolerate a renal biologic equivalent dose up to 40 Gy, on the basis of experience with radiopeptide treatment of neuroendocrine tumors (28). However, dosimetry is an important but not the only factor for determining the safety of a radionuclide treatment. As observed in a similar setting comparing somatostatin agonist and antagonist treatments, disproportionately higher hematotoxicity was observed with the somatostatin antagonist, with up to 57% of patients experiencing grade 4 hematotoxicity after 2 cycles (29).

High variability of absorbed doses was observed in tumor lesions, similar to data reported for 177Lu-PSMA I&T (5) and 177Lu-PSMA-617 (8,3032). The broad range might be partially attributable to the fact that our patients had only a few lesions. Similar to organ dosimetry, the relationship of the absorbed doses of both ligands is a more reliable parameter than the absolute values. Okamoto et al. and Baum et al. reported absorbed doses between 0.22 and 12.0 Gy/GBq and between 0.02 and 78 Gy/GBq, respectively, for 177Lu-PSMA I&T (5,33). For 177Lu-PSMA-617, Violet et al. reported mean absorbed doses of 5.28 Gy/GBq for bone metastases and 3.91 Gy/GBq for lymph node metastases (32).

Integrating all the previously discussed data for organs and tumor lesions, we calculated the TI for the kidneys in all patients (Fig. 3). Ultimately, 2 patients were treated with 177Lu-rhPSMA-7.3, as pretherapeutic dosimetry indicated a clear advantage. In 1 patient, 177Lu-PSMA I&T was used, given its clearly favorable profile. In the remaining 3 patients, the TI did not favor either of the 2 PSMA ligands. On the basis of the TI for the bone marrow, 3 patients had a favorable profile for 177Lu-rhPSMA-7.3 and 3 patients had a favorable profile for 177Lu-PSMA I&T.

Our analyses warrant some discussion on how the different characteristics of 177Lu-rhPSMA-7.3 can be exploited. Potential options for a drug development program would be the application of similar activities, as recommended for 177Lu-PSMA I&T and 177Lu-PSMA-617, which would lead to higher absorbed tumor doses and potential efficacy (34). This approach could be feasible given the so-far low toxicity profile of 177Lu-PSMA in general, with reported injected activities of up to 2 doses of 11 GBq of 177Lu-PSMA-617 (applied within 1 wk) (35). However, long-term toxicity for the kidneys of 177Lu-PSMA in general is unclear, and even the potential dose limits are controversial. Alternatively, similar tumor and organ doses with a smaller amount of activity and subsequently with a lower cost could be achieved, especially when non–carrier-added 177Lu is used for treatment (36). In the context of an expected large number of patients to be treated with PSMA RLT in the future, smaller amounts of 177Lu would also improve practical aspects (e.g., radioactive material program licensing, improved radiation safety for involved medical personnel).

Our retrospective analysis has limitations. First, only a small number of patients could be analyzed. Second, numerous factors can impair the accuracy of PET and planar dosimetry and can lead to a decreased correlation of the 2 modalities. Overlay in planar scintigraphy can lead to an overestimation of the absorbed dose, and further errors can occur for volumetric assessment (8,37). We tried to minimize such errors by adjusting the volume of interest using information from PET for the anatomic configuration of the lesions. However, especially for bone lesions, anatomic delineation can be difficult. Third, our dosimetry analyses for the bone marrow were prone to substantial challenges, as described earlier. As an alternative, we applied an additional method, using correction from tissue adjacent to the tissue in the thigh. In principle, this method is used in clinical dosimetry. However, as discussed earlier, it usually results in higher absorbed doses (less background to be subtracted in the thigh than adjacent to the lumbar spine), and no data are available in the literature to compare it with other PSMA ligands. Fourth, our pretherapeutic dosimetry using 1 GBq of 177Lu-rhPSMA-7.3 and 177Lu-PSMA I&T might already have achieved some therapeutic effect. Given the higher tumor doses delivered by 177Lu-rhPSMA-7.3, the dosimetry of subsequent 177Lu-PSMA I&T might be more affected than the dosimetry of 177Lu-rhPSMA-7.3 after 177Lu-PSMA I&T. We tried to minimize this bias by alternating the sequence of pretherapeutic applications.

CONCLUSION

Pretherapeutic clinical dosimetry confirmed preclinical results, with mean absorbed doses for tumors of 177Lu-rhPSMA-7.3 that were 2–3 times higher than those of 177Lu-PSMA I&T. Absorbed doses to normal organs increased at different levels, including the bone marrow. The newly introduced TI allowed for individual adjustment of absorbed tumor doses for the kidneys and the bone marrow as organs at risk. For the kidneys, it identified 2 of 6 patients with a clearly favorable biodistribution of 177Lu-rhPSMA-7.3 compared with 177Lu-PSMA I&T and a similar profile in 3 of 6 patients. For the bone marrow, a favorable profile was observed in 3 of 6 patients for 177Lu-rhPSMA-7.3 and in 3 of 6 patients for 177Lu-PSMA I&T. 177Lu-rhPSMA-7.3 holds promise for a therapeutic effect similar to that of 177Lu-PSMA I&T at lower absorbed doses and offers potential economical and radiation safety benefits.

DISCLOSURE

H.-J. Wester, A. Wurzer, and M. Eiber have applied for a patent for rhPSMA. H.-J. Wester is founder, shareholder, and advisory board member of Scintomics GmbH, Fuerstenfeldbruck, Germany. W.A. Weber reports prior consulting activities for Blue Earth Diagnostics Ltd. M. Eiber reports prior consulting activities for Blue Earth Diagnostics Ltd., Novartis, Telix, Progenics, Bayer, Point Biopharma, and Janssen. No other potential conflict of interest relevant to this article was reported.

KEY POINTS

QUESTION: Are the biodistribution, dosimetry, and therapeutic efficacy of 177Lu-rhPSMA-7.3 and 177Lu-PSMA I&T comparable?

PERTINENT FINDINGS: In mCRPC, pretherapeutic organ and tumor absorbed doses for 177Lu-rhPSMA-7.3 were higher than those for 177Lu-PSMA I&T, whereas the TI was equal to the mean for the kidney absorbed dose. Using 177Lu-rhPSMA-7.3 could lead to the same therapeutic effect without higher nephrotoxicity and with smaller amounts of radioactivity.

IMPLICATIONS FOR PATIENT CARE: Pretherapeutic data indicate higher tumor absorbed doses for 177Lu-rhPSMA-7.3 in radioligand treatment, a finding that should be explored in prospective clinical studies.

REFERENCES

  • 1. Lütje S, Heskamp S, Cornelissen AS, et al. PSMA ligands for radionuclide imaging and therapy of prostate cancer: clinical status. Theranostics. 2015;5:1388–1401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Heck MM, Tauber R, Schwaiger S, et al. Treatment outcome, toxicity, and predictive factors for radioligand therapy with 177Lu-PSMA-I&T in metastatic castration-resistant prostate cancer. Eur Urol. 2019;75:920–926. [DOI] [PubMed] [Google Scholar]
  • 3. Hofman MS, Violet J, Hicks RJ, et al. [177Lu]-PSMA-617 radionuclide treatment in patients with metastatic castration-resistant prostate cancer (LuPSMA trial): a single-centre, single-arm, phase 2 study. Lancet Oncol. 2018;19:825–833. [DOI] [PubMed] [Google Scholar]
  • 4. Morris MJ, De Bono JS, Chi KN, et al. Phase III study of lutetium-177-PSMA-617 in patients with metastatic castration-resistant prostate cancer (VISION) [abstract LBA4]. J Clin Oncol. 2021;39(suppl). [Google Scholar]
  • 5. Okamoto S, Thieme A, Allmann J, et al. Radiation dosimetry for 177Lu-PSMA I&T in metastatic castration-resistant prostate cancer: absorbed dose in normal organs and tumor lesions. J Nucl Med. 2017;58:445–450. [DOI] [PubMed] [Google Scholar]
  • 6. Kabasakal L, AbuQbeitah M, Aygun A, et al. Pre-therapeutic dosimetry of normal organs and tissues of 177Lu-PSMA-617 prostate-specific membrane antigen (PSMA) inhibitor in patients with castration-resistant prostate cancer. Eur J Nucl Med Mol Imaging. 2015;42:1976–1983. [DOI] [PubMed] [Google Scholar]
  • 7. Kratochwil C, Giesel FL, Stefanova M, et al. PSMA-targeted radionuclide therapy of metastatic castration-resistant prostate cancer with 177Lu-labeled PSMA-617. J Nucl Med. 2016;57:1170–1176. [DOI] [PubMed] [Google Scholar]
  • 8. Delker A, Fendler WP, Kratochwil C, et al. Dosimetry for 177Lu-DKFZ-PSMA-617: a new radiopharmaceutical for the treatment of metastatic prostate cancer. Eur J Nucl Med Mol Imaging. 2016;43:42–51. [DOI] [PubMed] [Google Scholar]
  • 9. Hohberg M, Eschner W, Schmidt M, et al. Lacrimal glands may represent organs at risk for radionuclide therapy of prostate cancer with [177Lu]DKFZ-PSMA-617. Mol Imaging Biol. 2016;18:437–445. [DOI] [PubMed] [Google Scholar]
  • 10. Kabasakal L, Toklu T, Yeyin N, et al. Lu-177-PSMA-617 prostate-specific membrane antigen inhibitor therapy in patients with castration-resistant prostate cancer: stability, bio-distribution and dosimetry. Mol Imaging Radionucl Ther. 2017;26:62–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Eiber M, Kroenke M, Wurzer A, et al. 18F-rhPSMA-7 PET for the detection of biochemical recurrence of prostate cancer after radical prostatectomy. J Nucl Med. 2020;61:696–701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Kroenke M, Wurzer A, Schwamborn K, et al. Histologically confirmed diagnostic efficacy of 18F-rhPSMA-7 PET for N-staging of patients with primary high-risk prostate cancer. J Nucl Med. 2020;61:710–715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Oh SW, Wurzer A, Teoh EJ, et al. Quantitative and qualitative analyses of biodistribution and PET image quality of a novel radiohybrid PSMA, 18F-rhPSMA-7, in patients with prostate cancer. J Nucl Med. 2020;61:702–709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Wurzer A, Di Carlo D, Schmidt A, et al. Radiohybrid ligands: a novel tracer concept exemplified by 18F- or 68Ga-labeled rhPSMA inhibitors. J Nucl Med. 2020;61:735–742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Wurzer A, Parzinger M, Konrad M, et al. Preclinical comparison of four [18F, (nat)Ga]rhPSMA-7 isomers: influence of the stereoconfiguration on pharmacokinetics. EJNMMI Res. 2020;10:149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Yusufi N, Wurzer A, Herz M, et al. Comparative preclinical biodistribution, dosimetry and endoradiotherapy in metastatic castration-resistant prostate cancer using 19F/177Lu-rhPSMA-7.3 and 177Lu-PSMA I&T. J Nucl Med. 2021;62:1106–1111. [DOI] [PubMed] [Google Scholar]
  • 17. O’Sullivan GJ, Carty FL, Cronin CG. Imaging of bone metastasis: an update. World J Radiol. 2015;7:202–211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Siegel JA, Thomas SR, Stubbs JB, et al. MIRD Pamphlet No. 16: techniques for quantitative radiopharmaceutical biodistribution data acquisition and analysis for use in human radiation dose estimates. J Nucl Med. 1999;40(suppl):37S–61S. [PubMed] [Google Scholar]
  • 19. Hindorf C, Glatting G, Chiesa C, Linden O, Flux G, Committee ED. EANM dosimetry committee guidelines for bone marrow and whole-body dosimetry. Eur J Nucl Med Mol Imaging. 2010;37:1238–1250. [DOI] [PubMed] [Google Scholar]
  • 20. Stabin MG, Sparks RB, Crowe E. OLINDA/EXM: the second-generation personal computer software for internal dose assessment in nuclear medicine. J Nucl Med. 2005;46:1023–1027. [PubMed] [Google Scholar]
  • 21. Blaickner M, Baum RP. Relevance of PET for pretherapeutic prediction of doses in peptide receptor radionuclide therapy. PET Clin. 2014;9:99–112. [DOI] [PubMed] [Google Scholar]
  • 22. Heck MM, Retz M, D’Alessandria C, et al. Systemic radioligand therapy with 177Lu labeled prostate specific membrane antigen ligand for imaging and therapy in patients with metastatic castration resistant prostate cancer. J Urol. 2016;196:382–391. [DOI] [PubMed] [Google Scholar]
  • 23. Gafita A, Fendler WP, Hui W, et al. Efficacy and safety of 177Lu-labeled prostate-specific membrane antigen radionuclide treatment in patients with diffuse bone marrow involvement: a multicenter retrospective study. Eur Urol. 2020;78:148–154. [DOI] [PubMed] [Google Scholar]
  • 24. Wester HJ, Schottelius M. PSMA-targeted radiopharmaceuticals for imaging and therapy. Semin Nucl Med. 2019;49:302–312. [DOI] [PubMed] [Google Scholar]
  • 25. Emami B, Lyman J, Brown A, et al. Tolerance of normal tissue to therapeutic irradiation. Int J Radiat Oncol Biol Phys. 1991;21:109–122. [DOI] [PubMed] [Google Scholar]
  • 26. Gallyamov M, Meyrick D, Barley J, Lenzo N. Renal outcomes of radioligand therapy: experience of 177lutetium–prostate-specific membrane antigen ligand therapy in metastatic castrate-resistant prostate cancer. Clin Kidney J. 2019;13:1049–1055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Dale R, Carabe-Fernandez A. The radiobiology of conventional radiotherapy and its application to radionuclide therapy. Cancer Biother Radiopharm. 2005;20:47–51. [DOI] [PubMed] [Google Scholar]
  • 28. Bodei L, Cremonesi M, Ferrari M, et al. Long-term evaluation of renal toxicity after peptide receptor radionuclide therapy with 90Y-DOTATOC and 177Lu-DOTATATE: the role of associated risk factors. Eur J Nucl Med Mol Imaging. 2008;35:1847–1856. [DOI] [PubMed] [Google Scholar]
  • 29. Reidy-Lagunes D, Pandit-Taskar N, O’Donoghue JA, et al. Phase I trial of well-differentiated neuroendocrine tumors (NETs) with radiolabeled somatostatin antagonist 177Lu-satoreotide tetraxetan. Clin Cancer Res. 2019;25:6939–6947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Fendler WP, Reinhardt S, Ilhan H, et al. Preliminary experience with dosimetry, response and patient reported outcome after 177Lu-PSMA-617 therapy for metastatic castration-resistant prostate cancer. Oncotarget. 2017;8:3581–3590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Scarpa L, Buxbaum S, Kendler D, et al. The 68Ga/177Lu theragnostic concept in PSMA targeting of castration-resistant prostate cancer: correlation of SUVmax values and absorbed dose estimates. Eur J Nucl Med Mol Imaging. 2017;44:788–800. [DOI] [PubMed] [Google Scholar]
  • 32. Violet J, Jackson P, Ferdinandus J, et al. Dosimetry of 177Lu-PSMA-617 in metastatic castration-resistant prostate cancer: correlations between pretherapeutic imaging and whole-body tumor dosimetry with treatment outcomes. J Nucl Med. 2019;60:517–523. [DOI] [PubMed] [Google Scholar]
  • 33. Baum RP, Kulkarni HR, Schuchardt C, et al. 177Lu-labeled prostate-specific membrane antigen radioligand therapy of metastatic castration-resistant prostate cancer: safety and efficacy. J Nucl Med. 2016;57:1006–1013. [DOI] [PubMed] [Google Scholar]
  • 34. Kratochwil C, Fendler WP, Eiber M, et al. EANM procedure guidelines for radionuclide therapy with 177Lu-labelled PSMA-ligands (177Lu-PSMA-RLT). Eur J Nucl Med Mol Imaging. 2019;46:2536–2544. [DOI] [PubMed] [Google Scholar]
  • 35. Tagawa ST, Niaz MJ, Osborne J, et al. Phase I/II dose-escalation trial of fractionated dose 177Lu-J591 plus 177Lu-PSMA-617 for metastatic castration-resistant prostate cancer (mCRPC) [abstract TPS339]. J Clin Oncol. 2019;37 (suppl). [Google Scholar]
  • 36. Tarasov VA, Andreev OI, Romanov EG, Kuznetsov RA, Kupriyanov VV, Tselishchev IV. Production of no-carrier added lutetium-177 by irradiation of enriched ytterbium-176. Curr Radiopharm. 2015;8:95–106. [DOI] [PubMed] [Google Scholar]
  • 37. Afshar-Oromieh A, Malcher A, Eder M, et al. PET imaging with a [68Ga]gallium-labelled PSMA ligand for the diagnosis of prostate cancer: biodistribution in humans and first evaluation of tumour lesions. Eur J Nucl Med Mol Imaging. 2013;40:486–495. [DOI] [PubMed] [Google Scholar]

Articles from Journal of Nuclear Medicine are provided here courtesy of Society of Nuclear Medicine and Molecular Imaging

RESOURCES