Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Jul 1.
Published in final edited form as: Eur Urol. 2021 Apr 8;80(1):82–94. doi: 10.1016/j.eururo.2021.03.004

A Systematic Review and Meta-analysis of the Effectiveness and Toxicities of Lutetium-177–labeled Prostate-specific Membrane Antigen–targeted Radioligand Therapy in Metastatic Castration-Resistant Prostate Cancer

Mohammad S Sadaghiani a, Sara Sheikhbahaei a, Rudolf A Werner a,b, Kenneth J Pienta c, Martin G Pomper a,c, Lilja B Solnes a, Michael A Gorin a,c, Nae-Yuh Wang d, Steven P Rowe a,c,*
PMCID: PMC8206006  NIHMSID: NIHMS1709621  PMID: 33840558

Abstract

Context:

Castration-resistant prostate cancer (CRPC) treatment is an evolving challenge. Prostate-specific membrane antigen (PSMA)-targeted endoradiotherapy/radioligand therapy (PRLT) with small-molecule, urea-based agents labeled with the β-particle–emitting radionuclide lutetium-177 (177Lu) is a promising new approach.

Objective:

In this systematic review and meta-analysis, we evaluated the efficacy and toxicity of PRLT.

Evidence acquisition:

A systematic search was performed in PubMed/Medline (last updated February 18, 2019). A total of 250 studies were reviewed, and 24 studies with 1192 patients were included in the analysis. Proportions of patients with ≥50% serum prostate-specific antigen (PSA) decrease, any PSA decrease, and any PSA increase were extracted. Proportions of patients showing any grade toxicity and those with grade 3/4 toxicities based on Common Terminology Criteria for Adverse Events (CTCAE) grading were extracted from manuscripts. Overall survival and progression-free survival were evaluated. A meta-analysis of single proportions was carried out. Furthermore, we compared the two most common PRLT agents, 177Lu-PSMA with 177Lu-PSMA-I&T, for effectiveness and toxicity.

Evidence synthesis:

Among the 24 included studies, 20 included data on 177Lu-PSMA-617, three included data on 177Lu-PSMA-I&T, and one study had aggregated data for 177Lu-PSMA-617 and 177Lu-PSMA-I&T. The estimated proportion of 177Lu-PSMA-617–treated patients who showed a serum PSA decrease of ≥50% with at least an 8-wk interval between therapy and PSA measurement was 0.44 (0.39; 0.50). Therapy with 177Lu-PSMA-I&T demonstrated an estimated proportion of patients with ≥50% PSA reduction to be 0.36 (0.26; 0.47). The aggregate results for men treated with more than one cycle of any kind of PRLT showed an estimated proportion of 0.46 (0.41; 0.51) for PSA response ≥50%. Regarding aggregate data from all of the PRLT agents, we found that grade 3 and 4 toxicities were uncommon, with estimated proportions from 0.01 (0.00;0.04) for nausea, fatigue, diarrhea, and elevated aspartate transaminase up to 0.08 (0.05; 0.12) for anemia. There was considerable heterogeneity among the studies in the “any-grade toxicity” groups. Meta-regression showed that more than one cycle of PRLT is associated with a greater proportion of patients with ≥50% PSA reduction. Overall survival according to pooled hazard ratios (HRs) for any PSA decline was 0.29 (0.18; 0.46), and for >50% PSA reduction was 0.67 (0.43; 1.07). Progression-free survival according to a pooled HR of >50% PSA reduction was 0.53 (0.32; 0.86).

Conclusions:

The relatively high number of PSA responders alongside the low rate of severe toxicity reflects the potentially promising role of PRLT in treating CRPC. The ultimate utility of this treatment modality will become clearer as multiple prospective studies continue to accrue. In the interim, this systematic review and meta-analysis can serve as a compendium of effectiveness and adverse events associated with PRLT for treating clinicians.

Patient summary:

Prostate-specific membrane antigen–targeted endoradiotherapy/radioligand therapy (PRLT) is associated with ≥50% reduction in prostate-specific antigen level in a large number of patients and a low rate of toxicity, reflecting its potential in treating castration-resistant prostate cancer. This systematic review and meta-analysis presents as a compendium of the effectiveness and adverse events related to PRLT for treating clinicians.

Keywords: Prostate cancer, Endoradiotherapy, Prostate-specific membrane antigen–targeted, endoradiotherapy/radioligand therapy

1. Introduction

Prostate cancer (PCa) is the second most common cancer in men worldwide, with an annual incidence of over 1.2 million and >350 000 deaths annually [1]. In the USA, PCa is the third most common cause of cancer death among men aged 60–79 yr and the second most common cause among men >80 yr of age [2]. A subgroup of patients show evidence of disease progression while on first-line systemic androgen deprivation therapy (ADT); this group of patients are considered to have castration-resistant prostate cancer (CRPC), a diagnosis that is associated with a high economic burden on patients and the healthcare system [3,4]. Treatment of these patients remains challenging, and new therapeutic approaches are being pursued actively [5]. Current recommendations include addition of taxane-based chemotherapeutic agents to ADT; however, this approach is associated with a significant increase in side effects [6]. Prostate-specific membrane antigen (PSMA)-targeted endoradiotherapy/radioligand therapy (PRLT) with small-molecule, urea-based agents labeled with the β-particle–emitting radionuclide lutetium-177 is a promising approach that has the potential to provide improved outcomes for patients with CRPC while also being well tolerated. There are multiple ongoing phase II and III clinical trials (ClinicalTrials.gov identifiers: NCT03511664, NCT03392428, and NCT03805594) as well as a large amount of retrospective data with PRLT [79].

PSMA is a transmembrane zinc metalloenzyme [10] that is highly expressed in the vast majority of PCa, including CRPC and poorly differentiated disease, and has a high internalization rate into PCa cells [11]. For these reasons, PSMA has become an attractive target for developing both diagnostic agents to image PCa and therapeutic agents to treat PCa. Additionally, 177Lu has favorable features that make it a good choice as a radionuclide for creating effective PRLT agents [12]. The emitted β-particles have limited tissue penetration of <2 mm, decreasing damage to healthy adjacent tissues. Lutetium-177 also emits low-energy gamma rays that can be utilized to image the tumor in real time during treatment. Its relatively long half-life of 6.73 d results in targeted cancers being exposed to the therapeutic effect for long periods of time. Different 177Lu-labeled PSMA-targeted ligands have been introduced, the two most common of which are the structurally similar agents 177Lu-PSMA-617 and 177Lu-PSMA-I&T (imaging and treatment). These agents can be considered in progressive CRPC after exhaustion of the approved lines of treatment [13], such as chemotherapy and newer antiandrogen agents [14], or in case of contraindication to the approved treatments [15].

As alluded to above, many of the reports on PRLT are retrospective evaluations in which endoradiotherapeutics were administered under the auspices of the “compassionate use” doctrine in Europe [16]. In addition to these retrospective studies [8], there has recently been an emergence of prospective studies from different parts of the world including Australia [15], India [17], and Austria [18]. These retrospective and prospective studies, outside of reporting on the rate of response of CRPC patients to PRLT, have provided a large volume of information on the toxicities associated with PRLT administration. Although prior systematic reviews and meta-analyses in this field have mostly focused on response to PRLT in patients with CRPC [1922], we reasoned that a more comprehensive overview of toxicities, and the severity of those toxicities, would serve as a useful reference for clinicians who refer patients, as well as nuclear medicine specialists who administer PRLT. Therefore, we conducted a comprehensive systematic review and meta-analysis of studies reporting Lu-labeled PSMA-targeted PRLT treatment outcomes with added focus on associated toxicities. In addition, we evaluated possible covariate contributions in the observed heterogeneity among studies, by conducting a meta-regression analysis.

2. Evidence acquisition

2.1. Literature review

This work was carried out according to the PICo method for clinical systematic reviews [23]. We searched PubMed/Medline for the following keywords: (177-Lu OR 177Lu OR Lu-177 OR Lutetium-177 OR theranostic OR theranostics) AND PSMA. A total of 250 entries were collected on February 18, 2019. All filters in PubMed were turned off, and there was no limit placed regarding the date of publication. The articles were reviewed against our inclusion and exclusion criteria: all retrospective or prospective studies of PRLT in humans, including single- or double-arm studies as well as randomized and nonrandomized trials were included in this meta-analysis. Only English-language articles were included. All studies evaluated a 177Lu-labeled, small-molecule PRLT ligand in patients with CRPC. For purposes of this analysis, treatment response was defined as a binary outcome of serum prostate-specific antigen (PSA) decline of ≥50%. We also collected the proportion of patients showing any level of PSA decrease as well as any level of PSA increase as additional binary outcomes. Among the studies that utilized PRLT in multiple cycles, when serum PSA alterations during the overall study time were available, the different cycle changes were ignored and only the overall response rate was included in the analysis [8,24]. When different sets of serum PSA levels were reported for different cycles in one article (without providing the overall study results), the highest number for each category (PSA decrease ≥50%, any PSA decrease, and PSA increase) was selected for a meta-analysis [2527].

Toxicities were extracted based on the Common Terminology Criteria for Adverse Events (CTCAE) grading system [28]. We categorized the toxicities into two groups: any-grade CTCAE toxicity and grade 3/4 CTCAE toxicities (ie, those with higher severity). Where applicable, we only included new toxicities that were attributed to the therapy and excluded baseline abnormalities. In papers where toxicities were not reported based on the CTCAE grading system, an estimate of the CTCAE grading was determined based on the description of the complication or further related information provided in the study whenever possible. If a toxicity was not provided based on the CTCAE grading system and no relevant clinical information was provided to allow us to generate an estimate of CTCAE grading, the number of patients manifesting that toxicity was included in the meta-analysis for all CTCAE grades, with the CTCAE grade 3/4 data blank for that study; for example, information regarding those who had fatigue was not provided in terms of CTCAE criteria in one manuscript [18]. Additional details regarding the included studies can be found in Table 1.

Table 1 – Summary of the studies included in the meta-analysis.

Number First author Year Country Study type PRLT agent Toxicity information included Number of patients treated Recruitment interval
1 Ahmadzadehfar [47] 2015 Germany Retrospective 177Lu-PSMA-617 Yes 10 November 2013 through January 2014
2 Ahmadzadehfar [48] 2017 Germany Retrospective 177Lu-PSMA-617 No 100 December 2014 through March 2017
3 Ahmadzadehfar [49] 2017 Germany Retrospective 177Lu-PSMA-617 Yes 49 NA
4 Ahmadzadehfar [27] 2017 Germany Retrospective 177Lu-PSMA-617 No 52 NA
5 Barber [9] 2019 Germany Retrospective 177Lu-PSMA-I&T and 177Lu-PSMA-617 Yes 132 March 2013 through December 2016
6 Brauer [26] 2017 Germany Retrospective 177Lu-PSMA-617 Yes 45 December 2014 through January 2017
7 Emmett [50] 2019 Australia Prospective 177Lu-PSMA-617 Yes 14 NA
8 Fendler [51] 2017 Germany Retrospective 177Lu-PSMA-617 Yes 15 September 2014 through May 2016
9 Ferdinandus [52] 2017 Germany Retrospective 177Lu-PSMA-617 No 40 NA
10 Grubmuller [53] 2019 Austria Retrospective 177Lu-PSMA-617 Yes 38 January 2015 through January 2018
11 Heck [7] 2019 Germany Retrospective 177Lu-PSMA-I&T Yes 100 December 2014 through August 2017
12 Hofman [15] 2018 Australia Prospective 177Lu-PSMA-617 Yes 30 August 2015 through December 2016
13 Kesavan [54] 2018 Australia Retrospective 177Lu-PSMA-I&T Yes 20 November 2015 through November 2016
14 Khurshid [55] 2018 Germany Retrospective 177Lu-PSMA-617 Yes 70 November 2014 through April 2016
15 Kletting [56] 2019 Germany Retrospective 177Lu-PSMA-I&T No 13 NA
16 Kratochwil [57] 2016 Germany Retrospective 177Lu-PSMA-617 Yes 30 NA
17 Rahbar [8] 2017 Germany Retrospective 177Lu-PSMA-617 Yes 145 February 2014 through July 2015
18 Rahbar [58] 2016 Germany Retrospective 177Lu-PSMA-617 Yes 74 December 2014 through November 2015
19 Rahbar [25] 2018 Germany Retrospective 177Lu-PSMA-617 No 71 December 2014 through November 2016
20 Rahbar [59] 2018 Germany Retrospective 177Lu-PSMA-617 No 104 December 2014 through December 2016
21 Rathke [60] 2018 Germany Retrospective 177Lu-PSMA-617 Yes 40 NA
22 Scarpa [18] 2017 Austria Prospective 177Lu-PSMA-617 Yes 10 NA
23 Yadav [17] 2017 India Prospective 177Lu-PSMA-617 Yes 31 2014–2016
24 Zang [46] 2019 China Prospective 177Lu-PSMA-617 Yes 5 November 2017 through February 2018

NA = not available; PRLT = PSMA-targeted endoradiotherapy/radioligand therapy; PSMA = prostate-specific membrane antigen.

Studies that are published in the same year by the same first author, are numbered to make comparison easier. The numbering is kept consistent during the whole manuscript including figures.

2.2. Meta-analysis

A meta-analysis of single proportions was performed for the proportion (95% confidence interval [CI]) of patients demonstrating a PSA decrease of ≥50%. The same approach was used to determine the estimated proportions of patients with any PSA response, as well as those who showed any increase in PSA. We determined the proportion of patients showing any toxicities based on the CTCAE criteria related to PSMA as well as proportion of patients showing adverse effects with CTCAE grade 3 or 4. All available results were gathered for 177Lu-PSMA-617 and 177Lu-PSMA-I&T, and the results were analyzed in aggregate as well as individually for 177Lu-PSMA-617 and 177Lu-PSMA-I&T (in retrospective studies in which more than one agent was utilized, the data for the individual agents were separated whenever possible). One study did not differentiate between the use of 177Lu-PSMA-617 and 177Lu-PSMA-I&T [9]; data from that study were utilized in the aggregated analysis only. Logit transformation with the inverse variance method was used to perform a meta-analysis of proportions. The inverse variance method was used because of the large number of studies with zero number of adverse effects, specifically for grade 3 or 4 toxicities. We performed the meta-analysis based on a random-effect model. I2 was calculated to quantify the heterogeneity. Forest plots were created with “meta” package in R. Moreover, PSA response and complications between 177Lu-PSMA-617 and 177Lu-PSMA-I&T were compared with SPSS version 25. Independent sample t test was performed to compare the mean of the outcome proportions of participants between 177Lu-PSMA-617 and 177Lu-PSMA-I&T.

2.3. Evaluation of bias

Funnel plots were used for the evaluation of publication bias [29].

2.4. Evaluation of heterogeneity

To evaluate the contribution of possible covariates in the heterogeneity, a meta-regression analysis was performed with “meta” package version 4.9–5 for study outcomes showing significant heterogeneity in the meta-analysis, with significant p value for χ2 test of heterogeneity. The following covariates were selected as possible contributing factors to the heterogeneity: prospective versus. retrospective studies, studies performed in economically developing countries versus developed countries, radioligand type (177Lu-PSMA-617 vs non–177Lu-PSMA-617), number of PRLT cycles (therapy with one cycle vs more than one cycle), and the time interval between PRLT and PSA evaluation (<8 vs ≥8 wk). To reduce the risk of data dredging, we performed meta-regression for variables that included more than eight studies. A p value of <0.05 was considered statistically significant. Proportions are reported, followed by 95% CIs in brackets.

In order to provide an idea of the diversity among prior therapies and also variables related to tumor burden, we provided the percentage of patients who received different types of therapies (prostatectomy, abiraterone, enzalutamide, chemotherapy including docetaxel or cabazitaxel, and 223Ra radiation therapy) and also the percentage of patients showing Gleason scores of ≤7, Gleason scores of >7, as well as local recurrence and metastasis status (lymph node, bone, and visceral, with detailed information regarding liver and lung wherever available) prior to PRLT in each study whenever available.

2.5. Evaluation of overall survival and progression-fee survival

The overall survival (OS) evaluation was based on the HRs of OS according to any PSA decline and a PSA decline of >50%. Progression-free survival (PFS) assessment was based on HRs according to >50% PSA decline. We extracted the estimated of HRs and 95% CI whenever provided by the authors. Regarding the studies that provided only Kaplan-Meier curves without readily available HR and 95% CI, GetData Graph Digitizer (http://www.getdata-graph-digitizer.com/) was used to read the graphical representations, and estimated HR and 95% CI were calculated based on a previous methodology [30]. A survival analysis was performed with review manager version 5.3 (The Cochrane Collaboration, Copenhagen, Norway). The rest of the meta-analysis was performed with R version 3.4.2 (2017-09-28) [31] and “meta” package version 4.9–5 [32].

3. Evidence synthesis

3.1. Results

3.1.1. Search results

A total of 250 articles were reviewed individually against the inclusion and exclusion criteria (Fig. 1). Twenty-nine articles met all the criteria. Five articles were removed from the meta-analysis because of an overlap of the analyzed patients with follow-up publications from the same institution [24,3336]. As such, a total of 24 studies including 1192 patients were used in the meta-analysis (Table 1); of these studies, 20 provided separate information for 177Lu-PSMA-617 (927 patients), three used 177Lu-PSMA-I&T (133 patients), and one (132 patients) used a mix of 177Lu-PSMA-617 and 177Lu-PSMA-I&T and reported aggregate data. None of the studies included a control group. Three studies were prospective [15,17,18], and the remaining studies were retrospective. The country where the study was conducted, the prospective versus retrospective approach, and toxicity information availability are included in Table 1. Patients in all studies had metastatic CRPC prior to PRLT, except for the study of Barber et al [9], where 39% of the patients were M0 and 37% were N0 prior to therapy.

Fig. 1 –

Fig. 1 –

Flow chart of the systematic review. PRISMA = Preferred Reporting Items for Systematic Review and Meta-analyses; PSA = prostate-specific antigen; PSMA = prostate-specific membrane antigen.

3.1.2. PSA response meta-analysis

Treatment response was determined based on ≥50% decline in serum PSA levels, in accordance to prior publications regarding PCa treatment [3739]. Figures 2 and 3 show the forest plots for the serum PSA response metrics within this meta-analysis.

Fig. 2 –

Fig. 2 –

Forest plots demonstrating the proportions of patients showing ≥50% decrease in PSA (A) after therapy with 177Lu-PSMA-I&T or 177Lu-PSMA-617, (B) after more than one cycle of therapy with 177Lu-PSMA-I&T or 177Lu-PSMA-617, (C) after therapy with 177Lu-PSMA-617, and (D) after therapy with 177Lu-PSMA-I&T. CI = confidence interval; df = degree of freedom; PSA = prostate-specific antigen; PSMA = prostate-specific membrane antigen; RLT = radioligand therapy.

Fig. 3 –

Fig. 3 –

Forest plots demonstrating the proportions of patients showing (A) any decrease in PSA after therapy with 177Lu-PSMA-I&T or 177Lu-PSMA-617, (B) any decrease in PSA after therapy with 177Lu-PSMA-617, and (C) any increase in PSA after therapy with 177Lu-PSMA-I&T or 177Lu-PSMA-617. CI = confidence interval; df = degree of freedom; PSA = prostate-specific antigen; PSMA = prostate-specific membrane antigen.

Among the studies that evaluated PSA ≥8 wk after the first PRLT, the estimated proportion of 177Lu-PSMA-617–treated patients who showed a serum PSA decrease of ≥50% was 0.44 (0.39; 0.50; Table 2). Those treated with 177Lu-PSMA-I&T showed an estimated proportion of 0.36 (0.26; 0.47) for a serum PSA decrease of ≥50%. No statistically significant difference was noted between response to 177Lu-PSMA-617 and 177Lu-PSMA-I&T. The details of the meta-regression analysis are provided under “Evaluation of heterogeneity” in the Results section.

Table 2 – Serum PSA alterations after PRLT.
PRLT ligand Serum PSA response metric No. of studies No. of patients Estimated proportion 95% Confidence interval I2 (%) p value (χ2 test of heterogeneity)
177Lu-PSMA-617 ≥50% PSA decrease (more than one cycle of PRLT) 16 657 0.47 0.41; 0.53 50 0.02
Any PSA decrease 16 783 0.70 0.66; 0.75 43 0.04
PSA increase 5 125 0.28 0.20; 0.36 0.0 0.82
177Lu-PSMA-I&T ≥ 50% PSA decrease 3 133 0.36 0.26; 0.47 18 0.29
177Lu-PSMA-I&T or 177Lu-PSMA-617 ≥ 50% PSA decrease 23 1161 0.41 0.36; 0.47 67 0 < 0.01
≥ 50% PSA decrease (only studies with ≥8 wk between PSA measurement and PRLT) 20 1073 0.44 0.40; 0.49 48 <0.01
Any PSA decrease 17 796 0.71 0.66; 0.75 42 0.03
177Lu-PSMA-I&T or 177Lu-PSMA-617 PSA increase 6 138 0.27 0.20; 0.35 0.0 0.79

PRLT = PSMA-targeted endoradiotherapy/radioligand therapy; PSA = prostate-specific antigen; PSMA = prostate-specific membrane antigen.

3.1.3. Toxicity meta-analysis

The proportion of patients who showed toxicities after PSMA therapy is shown in Table 3 as well as in Supplementary Figures 110.

Table 3 – Meta-analysis of the toxicities after PRLT.
Toxicity PRLT agent CTCAE grade 3 or 4 toxicity CTCAE any-grade toxicity
No. of studies, patients Estimated proportion 95% CI I2 (%) p value (χ2 test of heterogeneity) No. of studies, patients Estimated proportion 95% CI I2 p value (χ2 test of heterogeneity)
Anemia 177Lu-PSMA-I&T and 177Lu-PSMA-617 13, 718 0.08 0.05;0.12 42 0.06 11, 629 0.28 0.17; 0.42 88 <0.01
Diarrhea 177Lu-PSMA-I&T and 177Lu-PSMA-617 4, 244 0.01 0.00; 0.04 0.0 0.71 4, 244 0.06 0.02; 0.14 49 0.12
Elevated ALT 177Lu-PSMA-I&T and 177Lu-PSMA-617 4, 344 0.02 0.00; 0.10 55 0.08 4, 344 0.18 0.03; 0.62 96 <0.01
Elevated AST 177Lu-PSMA-I&T and 177Lu-PSMA-617 4, 45 0.01 0.0; 0.04 11 0.34 4, 345 0.21 0.12; 0.35 80 <0.01
Fatigue 177Lu-PSMA-I&T and 177Lu-PSMA-617 6,354 0.01 0.00; 0.04 0.0 0.68 7,364 0.25 0.16; 0.37 71 <0.01
Leukopenia 177Lu-PSMA-I&T and 177Lu-PSMA-617 13, 720 0.04 0.03; 0.07 13 0.31 10, 582 0.28 0.22; 0.35 61 <0.01
Nausea 177Lu-PSMA-I&T and 177Lu-PSMA-617 6, 423 0.01 0.00; 0.04 0.0 0.49 6, 423 0.12 0.04; 0.30 90 <0.01
Nephropathy 177Lu-PSMA-I&T and 177Lu-PSMA-617 8, 482 0.01 0.01; 0.04 0.0 0.66 8, 482 0.13 0.07; 0.24 78 <0.01
Thrombocytopenia 177Lu-PSMA-I&T and 177Lu-PSMA-617 14, 734 0.04 0.03; 0.06 0.0 0.66 12, 645 0.23 0.16; 0.31 72 <0.01
Xerostomia 177Lu-PSMA-I&T and 177Lu-PSMA-617 10, 488 0.02 0.01; 0.04 0.0 0.60 10, 488 0.22 0.12; 0.38 87 <0.01

ALT = alanine transaminase; AST = aspartate transaminase; CI = confidence interval; CTCAE = Common Terminology Criteria for Adverse Events; PRLT = PSMA-targeted endoradiotherapy/radioligandtherapy; PSMA = prostate-specific membrane antigen.

Comparison of toxicities between those who were treated with 177Lu-PSMA-617 and 177Lu-PSMA-I&T failed to show any significant difference. Overall, grade 3 and 4 toxicities were uncommon, with estimated proportions from 0.01 (0.00; 0.04) for nausea, fatigue, diarrhea, and elevated aspartate transaminase to 0.08 (0.05; 0.12) for anemia. Any-grade toxicities demonstrated considerable heterogeneity (Table 3).

In regard to CTCAE grade 3 or 4 toxicities, anemia was the highest reported complication among those who received 177Lu-PSMA-617 (0.19 [0.06; 0.15]) and 177Lu-PSMA-I&T (0.09 [0.05; 0.16]; Supplementary Table 1).

3.1.4. Survival meta-analysis

The pooled HR for the OS of any PSA decline was 0.29 (0.18–0.46) and for patients with >50% PSA decline was 0.67 (0.43–1.07). The pooled HR for the PFS of >50% PSA decline was 0.53 (0.32–0.86; Supplementary Fig. 11).

3.1.5. Comparison of 177Lu-PSMA-I&T and 177Lu-PSMA-617

A comparison of the proportion of patients showing ≥50% decline in serum PSA (p = 0.41, mean difference = 0.09, 95% CI [−1.3; 0.03]) or any decrease (p = 0.27, mean difference = −1.2, 95% CI [−0.3; −0.1]) in PSA between those who were treated with 177Lu-PSMA-I&T and those treated with 177Lu-PSMA-617 did not show any significant difference (for ≥50% decline variable: p = 0.41, mean difference = 0.09, 95% CI [−1.3; 0.03]; or for any PSA decrease: p = 0.27, mean difference = −1.2, 95% CI [−0.3; 0.1]). Furthermore, a comparison of the proportions of patients showing any-grade toxicity or grade 3 or 4 toxicities also did not show any significant difference between those who were treated with 177Lu-PSMA-617 and those who received 177Lu-PSMA-I&T (any-grade anemia: p = 0.98, mean difference = 0.005, 95% CI [−0.4; 0.04]; anemia with grade 3 or 4: p = 0.55, mean difference = −0.03, 95% CI [−0.13; 0.08]; any-grade diarrhea: p = 0.99, mean difference = 0.002, 95% CI [−0.48; 0.48]; any-grade fatigue: p = 0.56, mean difference = 0.09, 95% CI [−0.30; 0.48]; grade 3 or 4 fatigue: p = 0.70, mean difference = 0.003, 95% CI [−0.008; 0.01]; any-grade nausea: p = 0.48, mean difference = 0.17, 95% CI [−0.44; 0.79]; grade 3 or 4 nausea: p = 0.70, mean difference = 0.01, 95% CI [−0.08; 0.1]; any-grade thrombocytopenia: p = 0.95, mean difference = 0.007, 95% CI [−0.27; 0.29], grade 3 or 4 thrombocytopenia: p = 0.67, mean difference = −0.01, 95% CI [−0.08; 0.05]; grade 3 or 4 thrombocytopenia: p = 0.31, mean difference = −0.08, 95% CI [−0.58; 0.41]; grade 3 or 4 xerostomia: p = 0.64, mean difference = 0.01, 95% CI [−0.05; 0.07]; and any-grade xerostomia: p = 0.67, mean difference = 0.10, 95% CI [−0.40; 0.59]).

3.1.6. Evaluation of publication bias

Funnel plots (Supplementary Fig. 12) were used for the evaluation of possible publication bias. By visual inspection, there is no subjective asymmetry among the studies except for ≥50% PSA response, any-grade anemia, leukopenia, nephropathy, thrombocytopenia, and anemia with grade 3 or 4 toxicity.

3.1.7. Evaluation of heterogeneity

A meta-regression analysis of the proportion of patients with ≥50% PSA decline after radioligand therapy, regressed against the number of cycles of radioligand therapy (one cycle vs more than one cycle), demonstrated a significantly higher proportion of responded patients among those with more than one cycle of therapy (coefficient: 0.84, p < 0.001, 95% CI [0.36; 1.32]). Moreover, a meta-regression analysis of the proportion of patients with ≥50% PSA decline after radioligand therapy, regressed against the time interval between PRLT and PSA measurement (≥8 vs <8 wk interval) demonstrated a significant association of a higher proportion of responders with therapy in the ≥8-wk interval group (coefficient: 2.20, p < 0.001, 95% CI [1.30; 3.10]; Fig. 4A and 4B). The aggregate results for men treated with more than one cycle of PRLT (177Lu-PSMA-617 or 177Lu-PSMA-I&T) showed an estimated proportion of 0.46 (0.41; 0.51) for PSA response ≥50% (Fig. 2B).

Fig. 4 –

Fig. 4 –

Bubble plots demonstrating the proportions of patients with ≥50% PSA decline after radioligand therapy, regressed against (A) the time interval between PRLT and PSA measurement (<8- vs ≥8-wk interval) and (B) the cycles of radioligand therapy (therapy with one cycle vs more than one cycle). PRLT = PSMA-targeted endoradiotherapy/radioligand therapy; PSA = prostate-specific antigen; PSMA = prostate-specific membrane antigen.

The percentage of patients who received different types of therapies (including prostatectomy, abiraterone, enzalutamide, chemotherapy including docetaxel or cabazitaxel, and 223Ra radiation therapy) prior to PRLT and the percentage of patients showing Gleason scores of ≤7, Gleason scores of >7, local recurrence, and metastasis status (lymph node, bone, and visceral, with detailed information regarding liver and lung wherever available) prior to PRLT are provided in Supplementary Figures 13 and 14.

3.2. Discussion

In this systematic review and meta-analysis, we have provided a comprehensive evaluation of PSA status at the end of PRLT treatment and associated toxicity rates for patients with CRPC. The aggregate data showed that approximately 46% of CRPC patients being treated with more than one cycle of PRLT with either 177Lu-PSMA-I&T or 177Lu-PSMA-617 have PSA reductions of ≥50%, indicating that these agents are objectively effective for this patient population. The pooled estimated proportion of patients with grade 3 or 4 adverse events for all the evaluated toxicities was <10%. The relatively high number of PSA responders alongside the low rate of toxicity reflects the potentially promising role of PRLT in treating CRPC. Moreover, our study shows that a higher proportion of patients with ≥50% PSA reduction are associated with more than one cycle of PRLT in contrast to one cycle of therapy and also with a ≥8-wk interval between PRLT and PSA measurement in contrast to <8 wk.

To put the toxicities associated with PRLT in context, recent meta-analyses have evaluated the side effects with other treatment modalities for CRPC. For instance, Marchioni et al [40] showed that the rate of grade ≥3 adverse effects after abiraterone among chemotherapy-naïve patients with CRPC ranged from 4.4% to 15.5%. In another study, it was shown that early taxane-based chemotherapy with ADT might increase the risk of grade 3–5 adverse effects compared with ADT alone. According to the authors, addition of early taxane-based chemotherapy corresponded to 405 more grade 3–5 adverse events per 1000 (95% CI 243 more to 621 more) compared with those receiving ADT alone [6]. According to the CHAARTED trial, the range of grade 3/4 adverse events among patients who received the docetaxel-containing regimen was from 0.3% in thrombocytopenia to 12.1% in neutropenia [41]. Moreover, the STAMPEDE trial showed that grade 3–5 adverse events were reported among 32% of patients who received standard of care therapy, 32% of those who received standard of care therapy plus zoledronic acid, and 52% among those who received standard of care therapy plus docetaxel [42].

In a more recent meta-analysis with a focus on immunotherapy, the incidence of fatal adverse events was estimated as 3.2% [43]. Considering these numbers, the relatively low rate of severe toxicity with PRLT is quite promising. The most recent comparable systematic review about PSMA-targeted therapy in PCa is provided by Yadav et al [22]. Their meta-analysis included 17 studies with 744 patients based on their search done in August 2018. They provided estimated PSA response rates for 177Lu-PSMA-I&T and 177Lu-PSMA-617. The overall estimated proportion of patients showing any PSA decrease was 0.75 and that of patients with >50% decrease in PSA was 0.46. Both numbers are close to our estimate (0.41 for ≥50% decrease in PSA and 0.7 for any PSA decrease). Yadav et al [22] also provided descriptive results regarding toxicities from PSMA-targeted therapies, while in the current study we provided the forest plots and meta-analyzed results for ten complications.

The low number of severe adverse effects among patients who received PRLT agents may be the result of effective targeting of the cells that express PSMA. The expression of PSMA in non-PCa tissues would suggest candidate organs that may have off-target toxicities. These noncancerous tissues include the kidney, duodenum, and parotid and submandibular salivary glands [44]. Some of the adverse effects associated with PRLT agents based on this meta-analysis might be related to the abovementioned normal tissue sites of uptake. High uptake in the salivary glands may play a role in xerostomia. Normal liver uptake has been described as moderate [44], which may explain the relatively common elevation of transaminases. Anemia, leukopenia, and thrombocytopenia are perhaps more surprising given relatively low uptake in bone marrow; however, we believe that this effect is real as we subtracted the cases showing bone marrow suppression prior to PRLT whenever the data were provided.

There are other similar molecular structures utilized for PRLT that have been evaluated in a limited number of patients. For example, one study (ten patients) utilized 177Lu-iPSMA [45] and another study (four patients) used 177Lu-EB-PSMA [46]. Owing to limited data for these agents, we did not include these ligands in our meta-analysis. We also did not include α-particle–emitting agents in this meta-analysis, although, as data emerge with these compounds, a similar approach for the analysis efficacy and toxicities would be of value.

There are several limitations to this analysis. One limitation is the heterogeneity among the available studies; this includes the dose of radioligand provided, the time of PSA assessment, the interval between the injection and complication, and the diversity in prior treatments received for PCa.

Meta-regression was performed to find possible contributors to heterogeneity. At least part of the heterogeneity seen among studies for ≥50% PSA reduction can be explained by the difference in the number of cycles of the studies and the time interval between PRLT and PSA measurement. Our results showed that in contrast to studies with one cycle of PRLT, studies with more than one cycle are associated with a higher proportion of patients who responded to therapy. Moreover, there were a higher proportion of responders in studies with a ≥8-wk interval between PRLT and PSA measurement. A meta-analysis of the proportion of patients with ≥50% PSA reduction after more than one cycle of PRLT results in improved heterogeneity (I2 decrease from 67% to 51%). Moreover, limiting the studies to those with at least an 8-wk interval between PRLT and PSA evaluation resulted in improved heterogeneity (I2 decreased from 70% to 52%). We noticed considerable heterogeneity among studies for any-grade toxicities. The meta-regression failed to show the evaluated factors, namely, retrospective versus prospective studies, the economic situation of the countries, and the specific type of therapeutic agent (177Lu-PSMA-617 vs 177Lu-PSMA-I&T) as a contributing factor. However, a possible explanation can be the different time intervals of reporting the complication, although many studies did not provide the interval between noticing the toxicity and PRLT initiation. The other possible contributor to heterogeneity is prior therapies, including chemotherapies and new antiandrogen agents that were distributed diversely across studies. The tumor burden prior to therapy is another possible contributor to the heterogeneity. Variables such as Gleason score and metastatic status may affect the way patients responded to PRLT. The details of the proportion of patients with different therapies and burden scores prior to PRLT are provided in Supplementary Figures 13 and 14, and Supplementary Tables 3 and 4.

There are intrinsic limitations to the single-arm designs of the studies that underpin our findings. All studies were single arm; as a result, none of the included studies was randomized. Promisingly, among the studies that have been included in this meta-analysis, three were prospective studies [1518]. According to ClinicalTrials.gov, there are 15 ongoing trials examining the role of 177Lu-labeled PSMA-targeted agents in CRPC, among which three are double-arm studies. The control group in a multicenter study, which is recruiting 750 patients from several countries in North America and Europe, receives the best supportive/standard of care therapy, in contrast to the trial group that is being treated with 177Lu-PSMA-617. The study is expected to be completed by May 2021 (ClinicalTrials.gov identifier: NCT03511664). There is another major ongoing trial from Australia that compares 177Lu-PSMA-617 with cabazitaxel in a randomized study with 200 patients and is expected to be completed by October 2021 (ClinicalTrials.gov identifier: NCT03392428). Moreover, there is a nonrandomized sequential study of 177Lu-PSMA-617 with pembrolizumab as an immunotherapeutic agent that is estimated to start in May 2019 (ClinicalTrials.gov identifier: NCT03805594). The results of this meta-analysis provide a comprehensive overview of the available data regarding the effectiveness and toxicities related to PRLT in patients with CRPC, and can be integral to clinician decision-making and appropriate counseling of patients until the results of the ongoing trials become available and we can further refine our under-standing of this evolving field.

4. Conclusions

The relatively high number of PSA responders alongside the low rate of severe toxicity indicates the potentially promising role of PRLT in treating CRPC. This systematic review and meta-analysis can serve as a compendium of effectiveness and adverse events for treating clinicians to balance the risks and benefits of this novel therapy for CRPC.

Supplementary Material

Supplementary material

Acknowledgments:

We are indebted to Dr. Bruce J. Trock for helpful discussion regarding the analysis performed in this manuscript.

Funding/Support and role of the sponsor:

This publication was supported by the National Institute of Biomedical Imaging and Bioengineering of the National Institutes of Health under award number T32EB006351. Moreover, we would like to acknowledge the support for the statistical analysis by the National Center for Research Resources and the National Center for Advancing Translational Sciences (NCATS) of the National Institutes of Health through grant number 1UL1TR001079. We also acknowledge funding from CA134675, CA183031, CA184228, EB024495, the Prostate Cancer Foundation Young Investigator Award, and the European Union’s Horizon 2020 research and innovation program under Marie Sklodowska-Curie grant agreement 701983.

Financial disclosures:

Steven P. Rowe certifies that all conflicts of interest, including specific financial interests and relationships and affiliations relevant to the subject matter or materials discussed in the manuscript (eg, employment/affiliation, grants or funding, consultancies, honoraria, stock ownership or options, expert testimony, royalties, or patents filed, received, or pending), are the following: None.

Footnotes

Appendix A. Supplementary data

Supplementary material related to this article can be found, in the online version, at doi:https://doi.org/10.1016/j.eururo.2021.03.004.

References

  • [1].Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018;68:394–424. [DOI] [PubMed] [Google Scholar]
  • [2].Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin 2019;69:7–34. [DOI] [PubMed] [Google Scholar]
  • [3].Satoh T, Ledesma D, Yoshihara N. The economic burden of metastatic castration resistant prostate cancer and skeletal related events in Japanese university hospitals. Asian Pac J Cancer Prev APJCP 2018;19:21–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Grochtdreis T, Konig HH, Dobruschkin A, von Amsberg G, Dams J. Cost-effectiveness analyses and cost analyses in castration-resistant prostate cancer: a systematic review. PLoS One 2018;13: e0208063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Sumanasuriya S, De Bono J. Treatment of advanced prostate cancer—a review of current therapies and future promise. Cold Spring Harb Perspect Med 2018;8:a030635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Sathianathen NJ, Philippou YA, Kuntz GM, et al. Taxane-based chemohormonal therapy for metastatic hormone-sensitive prostate cancer. Cochrane Database Syst Rev 2018;10:CD012816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].Heck MM, Tauber R, Schwaiger S, et al. Treatment outcome, toxicity, and predictive factors for radioligand therapy with (177)Lu-PSMA-I&T in metastatic castration-resistant prostate cancer. Eur Urol 2019;75:920–6. [DOI] [PubMed] [Google Scholar]
  • [8].Rahbar K, Ahmadzadehfar H, Kratochwil C, et al. German multicenter study investigating 177Lu-PSMA-617 radioligand therapy in advanced prostate cancer patients. J Nucl Med 2017;58:85–90. [DOI] [PubMed] [Google Scholar]
  • [9].Barber TW, Singh A, Kulkarni HR, Niepsch K, Billah B, Baum RP. Clinical outcomes of (177)Lu-PSMA radioligand therapy in taxane chemotherapy pretreated and taxane chemotherapy naive patients with metastatic castration resistant prostate cancer. J Nucl Med 2019;60:955–62. [DOI] [PubMed] [Google Scholar]
  • [10].Mesters JR, Barinka C, Li W, et al. Structure of glutamate carboxy-peptidase II, a drug target in neuronal damage and prostate cancer. EMBO J 2006;25:1375–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].von Eyben FE, Baumann GS, Baum RP. PSMA diagnostics and treatments of prostate cancer become mature. Clin Transl Imaging 2018;6:145–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Emmett L, Willowson K, Violet J, Shin J, Blanksby A, Lee J. Lutetium (177) PSMA radionuclide therapy for men with prostate cancer: a review of the current literature and discussion of practical aspects of therapy. J Med Radiat Sci 2017;64:52–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Fendler WP, Rahbar K, Herrmann K, Kratochwil C, Eiber M. (177)Lu-PSMA Radioligand therapy for prostate cancer. J Nucl Med 2017;58:1196–200. [DOI] [PubMed] [Google Scholar]
  • [14].Ferriero M, Mastroianni R, De Nunzio C, et al. Managing lines of therapy in castration-resistant prostate cancer: real-life snapshot from a multicenter cohort. World J Urol 2020;38:1757–64. [DOI] [PubMed] [Google Scholar]
  • [15].Hofman MS, Violet J, Hicks RJ, et al. [(177)Lu]-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;19:825–33. [DOI] [PubMed] [Google Scholar]
  • [16].Rahbari M, Rahbari NN. Compassionate use of medicinal products in Europe: current status and perspectives. Bull World Health Org 2011;89:163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Yadav MP, Ballal S, Tripathi M, et al. (177)Lu-DKFZ-PSMA-617 therapy in metastatic castration resistant prostate cancer: safety, efficacy, and quality of life assessment. Eur J Nucl Med Mol Imaging 2017;44:81–91. [DOI] [PubMed] [Google Scholar]
  • [18].Scarpa L, Buxbaum S, Kendler D, et al. The (68)Ga/(177)Lu 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]
  • [19].Calopedos RJS, Chalasani V, Asher R, Emmett L, Woo HH. Lutetium-177-labelled anti-prostate-specific membrane antigen antibody and ligands for the treatment of metastatic castrate-resistant prostate cancer: a systematic review and meta-analysis. Prostate Cancer Prostatic Dis 2017;20:352–60. [DOI] [PubMed] [Google Scholar]
  • [20].von Eyben FE, Roviello G, Kiljunen T, et al. Third-line treatment and (177)Lu-PSMA radioligand therapy of metastatic castration-resistant prostate cancer: a systematic review. Eur J Nucl Med Mol Imaging 2018;45:496–508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Kim YJ, Kim YI. Therapeutic responses and survival effects of 177Lu-PSMA-617 radioligand therapy in metastatic castrate-resistant prostate cancer: a meta-analysis. Clin Nucl Med 2018;43:728–34. [DOI] [PubMed] [Google Scholar]
  • [22].Yadav MP, Ballal S, Sahoo RK, Dwivedi SN, Bal C. Radioligand therapy with (177)Lu-PSMA for metastatic castration-resistant prostate cancer: a systematic review and meta-analysis. AJR Am J Roentgenol 2019;213:275–85. [DOI] [PubMed] [Google Scholar]
  • [23].Schardt C, Adams MB, Owens T, Keitz S, Fontelo P. Utilization of the PICO framework to improve searching PubMed for clinical questions. BMC Med Inform Decis Mak 2007;7:16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Heck MM, Retz M, D’Alessandria C, et al. Systemic radioligand therapy with (177)Lu labeled prostate specific membrane antigen ligand for imaging and therapy in patients with metastatic castration resistant prostate cancer. J Urol 2016;196:382–91. [DOI] [PubMed] [Google Scholar]
  • [25].Rahbar K, Bogeman M, Yordanova A, et al. Delayed response after repeated (177)Lu-PSMA-617 radioligand therapy in patients with metastatic castration resistant prostate cancer. Eur J Nucl Med Mol Imaging 2018;45:243–6. [DOI] [PubMed] [Google Scholar]
  • [26].Brauer A, Grubert LS, Roll W, et al. (177)Lu-PSMA-617 radioligand therapy and outcome in patients with metastasized castration-resistant prostate cancer. Eur J Nucl Med Mol Imaging 2017;44:1663–70. [DOI] [PubMed] [Google Scholar]
  • [27].Ahmadzadehfar H, Wegen S, Yordanova A, et al. Overall survival and response pattern of castration-resistant metastatic prostate cancer to multiple cycles of radioligand therapy using [(177)Lu]Lu-PSMA-617. Eur J Nucl Med Mol Imaging 2017;44:1448–54. [DOI] [PubMed] [Google Scholar]
  • [28].National Cancer Institute. Common Terminology Criteria for Adverse Events (CTCAE).
  • [29].Sterne JA, Sutton AJ, Ioannidis JP, et al. Recommendations for examining and interpreting funnel plot asymmetry in meta-analyses of randomised controlled trials. BMJ 2011;343:d4002. [DOI] [PubMed] [Google Scholar]
  • [30].Tierney JF, Stewart LA, Ghersi D, Burdett S, Sydes MR. Practical methods for incorporating summary time-to-event data into meta-analysis. Trials 2007;8:16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Team RC. R: A Language and environment for statistical computing. 3.4.2 ed Vienna, Austria: R Foundation for Statistical Computing; 2017. [Google Scholar]
  • [32].Guido S meta: An R package for meta-analysis. R News 2007;7:40–5. [Google Scholar]
  • [33].Kulkarni HR, Singh A, Schuchardt C, et al. PSMA-based radioligand therapy for metastatic castration-resistant prostate cancer: the bad Berka experience since 2013. J Nucl Med 2016;57:97S–104S. [DOI] [PubMed] [Google Scholar]
  • [34].Rahbar K, Bode A, Weckesser M, et al. Radioligand therapy with 177Lu-PSMA-617 as a novel therapeutic option in patients with metastatic castration resistant prostate cancer. Clin Nucl Med 2016;41:522–8. [DOI] [PubMed] [Google Scholar]
  • [35].Ahmadzadehfar H, Eppard E, Kurpig S, et al. Therapeutic response and side effects of repeated radioligand therapy with 177Lu-PSMA-DKFZ-617 of castrate-resistant metastatic prostate cancer. Oncotarget 2016;7:12477–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].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;57:1006–13. [DOI] [PubMed] [Google Scholar]
  • [37].Ryan CJ, Smith MR, Fizazi K, et al. Abiraterone acetate plus prednisone versus placebo plus prednisone in chemotherapy-naive men with metastatic castration-resistant prostate cancer (COU-AA-302): final overall survival analysis of a randomised, double-blind, placebo-controlled phase 3 study. Lancet Oncol 2015;16:152–60. [DOI] [PubMed] [Google Scholar]
  • [38].Scher HI, Fizazi K, Saad F, et al. Increased survival with enzalutamide in prostate cancer after chemotherapy. N Engl J Med 2012;367:1187–97. [DOI] [PubMed] [Google Scholar]
  • [39].Petrylak DP, Tangen CM, Hussain MH, et al. Docetaxel and estramustine compared with mitoxantrone and prednisone for advanced refractory prostate cancer. N Engl J Med 2004;351:1513–20. [DOI] [PubMed] [Google Scholar]
  • [40].Marchioni M, Sountoulides P, Bada M, et al. Abiraterone in chemotherapy-naive patients with metastatic castration-resistant prostate cancer: a systematic review of’ real-life’ studies. Ther Adv Urol 2018;10:305–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Sweeney CJ, Chen YH, Carducci M, et al. Chemohormonal therapy in metastatic hormone-sensitive prostate cancer. N Engl J Med 2015;373:737–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].James ND, Sydes MR, Clarke NW, et al. Addition of docetaxel, zoledronic acid, or both to first-line long-term hormone therapy in prostate cancer (STAMPEDE): survival results from an adaptive, multiarm, multistage, platform randomised controlled trial. Lancet 2016;387:1163–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Jiang Y, Zhang N, Pang H, Gao X, Zhang H. Risk and incidence of fatal adverse events associated with immune checkpoint inhibitors: a systematic review and meta-analysis. Ther Clin Risk Manag 2019;15:293–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Sheikhbahaei S, Afshar-Oromieh A, Eiber M, et al. Pearls and pitfalls in clinical interpretation of prostate-specific membrane antigen (PSMA)-targeted PET imaging. Eur J Nucl Med Mol Imaging 2017;44:2117–36. [DOI] [PubMed] [Google Scholar]
  • [45].Santos-Cuevas C, Ferro-Flores G, Garcia-Perez FO, et al. (177)Lu-DOTA-HYNIC-Lys(Nal)-Urea-Glu: biokinetics, dosimetry, and evaluation in patients with advanced prostate cancer. Contrast Media Mol Imaging 2018;2018:5247153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Zang J, Fan X, Wang H, et al. First-in-human study of (177)Lu-EB-PSMA-617 in patients with metastatic castration-resistant prostate cancer. Eur J Nucl Med Mol Imaging 2019;46:148–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Ahmadzadehfar H, Rahbar K, Kurpig S, et al. Early side effects and first results of radioligand therapy with (177)Lu-DKFZ-617 PSMA of castrate-resistant metastatic prostate cancer: a two-centre study. EJNMMI Res 2015;5:114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Ahmadzadehfar H, Schlolaut S, Fimmers R, et al. Predictors of overall survival in metastatic castration-resistant prostate cancer patients receiving [(177)Lu]Lu-PSMA-617 radioligand therapy. Oncotarget 2017;8:103108–1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Ahmadzadehfar H, Zimbelmann S, Yordanova A, et al. Radioligand therapy of metastatic prostate cancer using (177)Lu-PSMA-617 after radiation exposure to (223)Ra-dichloride. Oncotarget 2017;8:55567–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Emmett L, Crumbaker M, Ho B, et al. Results of a prospective phase 2 pilot trial of (177)Lu-PSMA-617 therapy for metastatic castration-resistant prostate cancer including imaging predictors of treatment response and patterns of progression. Clin Genitourin Cancer 2019;17:15–22. [DOI] [PubMed] [Google Scholar]
  • [51].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 can-cer. Oncotarget 2017;8:3581–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Ferdinandus J, Eppard E, Gaertner FC, et al. Predictors of response to radioligand therapy of metastatic castrate-resistant prostate cancer with 177Lu-PSMA-617. J Nucl Med 2017;58:312–9. [DOI] [PubMed] [Google Scholar]
  • [53].Grubmuller B, Senn D, Kramer G, et al. Response assessment using (68)Ga-PSMA ligand PET in patients undergoing (177)Lu-PSMA radioligand therapy for metastatic castration-resistant prostate cancer. Eur J Nucl Med Mol Imaging 2019;46: 1063–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Kesavan M, Turner JH, Meyrick D, Yeo S, Cardaci G, Lenzo NP. Salvage radiopeptide therapy of advanced castrate-resistant prostate cancer with lutetium-177-labeled prostate-specific membrane antigen: efficacy and safety in routine practice. Cancer Biother Radiopharm 2018;33:274–81. [DOI] [PubMed] [Google Scholar]
  • [55].Khurshid Z, Ahmadzadehfar H, Gaertner Fc, et al. Role of textural heterogeneity parameters in patient selection for 177Lu-PSMA therapy via response prediction. Oncotarget 2018;9:33312–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Kletting P, Thieme A, Eberhardt N, et al. Modeling and predicting tumor response in radioligand therapy. J Nucl Med 2019;60:65–70. [DOI] [PubMed] [Google Scholar]
  • [57].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;57:1170–6. [DOI] [PubMed] [Google Scholar]
  • [58].Rahbar K, Schmidt M, Heinzel A, et al. Response and tolerability of a single dose of 177Lu-PSMA-617 in patients with metastatic castration-resistant prostate cancer: a multicenter retrospective analysis. J Nucl Med 2016;57:1334–8. [DOI] [PubMed] [Google Scholar]
  • [59].Rahbar K, Boegemann M, Yordanova A, et al. PSMA targeted radioligand therapy in metastatic castration resistant prostate cancer after chemotherapy, abiraterone and/or enzalutamide. A retrospective analysis of overall survival. Eur J Nucl Med Mol Imaging 2018;45:12–9. [DOI] [PubMed] [Google Scholar]
  • [60].Rathke H, Giesel FL, Flechsig P, et al. Repeated (177)Lu-labeled PSMA-617 radioligand therapy using treatment activities of up to 9.3 GBq. J Nucl Med 2018;59:459–65. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary material

RESOURCES