Purpose:

The anti-CD33 antibody lintuzumab has modest activity against acute myeloid leukemia (AML). To increase its potency, lintuzumab was conjugated to actinium-225 (225Ac), a radionuclide yielding 4 α-particles. This first-in-human, phase I trial was conducted to determine the safety, pharmacology, and biological activity of 225Ac-lintuzumab.

Patients and Methods:

Eighteen patients (median age, 64 years; range, 45–80) with relapsed or refractory AML received a single infusion of 225Ac-lintuzumab at activities of 18.5 to 148 kBq/kg.

Results:

The maximum tolerated dose was 111 kBq/kg. Dose-limiting toxicities included myelosuppression lasting > 35 days in one patient receiving 148 kBq/kg and death from sepsis in two patients treated with 111 and 148 kBq/kg. Myelosuppression was the most common toxicity. Significant extramedullary toxicities were limited to transient grade 3 liver function abnormalities. Pharmacokinetics were determined by gamma counting serial whole blood, plasma, and urine samples at energy windows for the 225Ac daughters, francium-221 and bismuth-213. Two-phase elimination kinetics were seen with mean plasma t1/2 − α and t1/2 − β of 1.9 and 38 hours, respectively. Peripheral blood blasts were eliminated in 10 of 16 evaluable patients (63%) but only at doses of ≥ 37 kBq/kg. Bone marrow blasts were reduced in 10 of 15 evaluable patients (67%), including 3 patients with marrow blasts ≤ 5% and one patient with a morphologic leukemia-free state.

Conclusions:

Therapy for AML with the targeted α-particle generator 225Ac-lintuzumab was feasible with an acceptable safety profile. Elimination of circulating blasts or reductions in marrow blasts were observed across all dose levels.

Translational Relevance

This is the first study to show that targeted therapy with the in vivo α-particle generator 225Ac-lintuzumab is feasible, has an acceptable safety profile, and has activity against advanced acute myeloid leukemia. It provides the rationale for the development of 225Ac-lintuzumab in combination with cytotoxic chemotherapy and other targeted agents, for elimination of measurable residual disease, as conditioning for hematopoietic cell transplantation for AML and myelodysplastic syndromes, and in other CD33-expressing malignancies. As myelosuppression is dose limiting, additional phase I studies will be needed as this novel treatment modality is integrated into treatment programs with other agents and used in various clinical settings.

Acute myeloid leukemia (AML) is a cytogenetically and molecularly heterogeneous hematologic neoplasm characterized by lack of differentiation and increased proliferation of early myeloid progenitor cells. Data from the Surveillance, Epidemiology, and End Results (SEER) program of the NCI report 5-year survival rates of 47.5% and 8.2% for patients younger than 65 years and 65 years or older, respectively (1). Despite the U.S. Food and Drug Administration approval of nine new agents for AML since 2017, novel agents are still needed to improve long-term outcomes in this difficult-to-cure disease.

Lintuzumab (HuM195) is a humanized monoclonal antibody that targets CD33, a 67-kDa cell-surface glycoprotein expressed on most myeloid leukemia cells. Previous studies demonstrated that lintuzumab can target leukemia cells in patients without eliciting immunogenicity (2), can eliminate minimal residual disease in acute promyelocytic leukemia (3), and can produce occasional remissions in AML (4–6). When added to standard chemotherapy, however, unconjugated lintuzumab was unable to improve response rate or survival in two randomized trials (7, 8). Therefore, to increase its modest single-agent activity in AML, lintuzumab was initially conjugated to the β particle-emitting isotopes iodine-131 or yttrium-90. Studies with these agents demonstrated that β particle-emitting conjugates of anti-CD33 constructs could eliminate large leukemic burdens but required hematopoietic cell transplantation (HCT) due to profound myelosuppression (9, 10).

In the effort to enhance tumor cell killing and simultaneously reduce the nonspecific cytotoxic effects seen with β-emitters, investigation of α-particle therapy was undertaken. Compared with β-particles, α-particles have a shorter range (50–80 vs. 800–10,000 µm) and a higher linear energy transfer (100 vs. 0.2 keV/µm; ref. 11). As few as 1 or 2 α-particles can kill a target cell. We have previously reported our experience with the α-emitter conjugate bismuth-213 (213Bi)-lintuzumab, which demonstrated significant antileukemic activity, including complete responses following cytoreduction in AML (12, 13). However, the widespread clinical utility of radioimmunotherapy with 213Bi is limited by its short 46-minute half-life and the need for an onsite 225Ac/213Bi generator.

225Ac is an isotope of actinium with a half-life of 10 days. In its decay to stable 209Bi, 225Ac generates francium-221 (221Fr), astatine-217 (217At), 213Bi, and lead-209 (209Pb), yielding 4 α-particles (Fig. 1). 225Ac can be conjugated to various antibodies using the bifunctional chelate p-isothiocyanatobenzyl-1,4,7,10-tetraaczacyclododecane-1,4,7,10-tetraacetic acid (DOTA-SCN). One solution to the limitations of 213Bi is to use lintuzumab as a vehicle to deliver 225Ac to the target leukemia cell. In this way, 225Ac serves as an in vivo α-particle generator, such that the decay produces a cascade of 4 α emissions. 225Ac-labeled immunoconjugates kill in vitro at radioactivity doses at least 1,000 times lower than 213Bi analogues and prolong survival in mouse xenograft models of several cancers (14). Treatment with 225Ac-labeled prostate membrane-specific antigen (PSMA) ligand has shown significant activity in advanced, metastatic prostate carcinoma (15, 16). To determine the maximum-tolerated dose (MTD), safety, pharmacology, and biological activity of 225Ac-lintuzumab in AML, we conducted a first-in-human, phase I dose-escalation trial.

Figure 1.

225Ac decay scheme. 225Ac produces six predominant radionuclide daughters in the decay cascade to stable 209Bi. A single Ac decay yields net 4 alpha and 3 beta disintegrations, most of high energy, and 2 useful gamma emissions.

Figure 1.

225Ac decay scheme. 225Ac produces six predominant radionuclide daughters in the decay cascade to stable 209Bi. A single Ac decay yields net 4 alpha and 3 beta disintegrations, most of high energy, and 2 useful gamma emissions.

Close modal

225Ac-lintuzumab preparation

225Ac-lintuzumab is a radioimmunoconjugate composed of humanized anti-CD33 monoclonal antibody lintuzumab (Protein Design Labs, Inc.) linked to 225Ac, via the bifunctional chelate DOTA-SCN. 225Ac, supplied by Actinium Pharmaceuticals, was obtained from Oak Ridge National Laboratory. 225Ac was conjugated to lintuzumab using a 2-step labeling method (17). First, 225Ac was labeled to DOTA-SCN (Macrocyclics and Dow Chemical Company) species at pH 4.5 to 5 in acetate buffer at 55°C to 60°C for 30 minutes. Then, 225Ac-DOTA-SCN was mixed with lintuzumab in carbonate buffer at pH 8.5 to 9 at 37°C for 30 minutes. The final product was purified by size exclusion chromatography using a 10-mL Bio-Rad 10DG column and 1% human serum albumin (HSA). Typical reaction yields were 10% ± 5%, which were sufficient for this study. Antibody doses were adjusted to maintain specific activity levels of 1.23 to 5.92 kBq/μg. 225Ac activity and antibody mass were adjusted per body weight: < 74 kBq/kg were labeled on 15 µg/kg of lintuzumab. 225Ac activities 74 and 111 kBq/kg were labeled on 20 µg/kg of lintuzumab. 225Ac activities of 148 kBq/kg were labeled on 25 µg/kg of lintuzumab. Filtered 225Ac-lintuzumab was diluted in 50-mL normal saline with 1% HSA for intravenous (i.v.) infusion through an online 0.2-µm filter. The final product was infused i.v. over 15 to 30 minutes using an infusion pump.

Patient eligibility

Eligible patients included those in three restricted categories of AML, chronic myeloid leukemia (CML), or myelodysplastic syndromes (MDS). Eligible patients with AML had relapsed AML or AML refractory to 2 courses of standard induction chemotherapy or to 1 course of high-dose cytarabine-based induction chemotherapy. Eligible patients with CML were in accelerated phase or myeloid blast crisis progressing after imatinib and a second-generation tyrosine kinase inhibitor. Eligible patients with MDS had MDS with excess blasts or chronic myelomonocytic leukemia with International Prognostic Scoring System (IPSS) score ≥ 2.5 and were refractory to or relapsed after a hypomethylating agent. More than 25% of the patient's bone marrow blasts were required to express CD33. No antileukemic therapy was administered for 3 weeks before study entry except for hydroxyurea, which was discontinued before treatment with 225Ac-lintuzumab. Concurrent use of either oral or i.v. antibiotics was allowed. Entry criteria included adequate renal and hepatic function as demonstrated by a serum creatinine ≤ 1.5 mg/dL, a calculated creatinine clearance > 60 mL/min, < 1 g urinary protein/24 hours, bilirubin ≤1.5 mg/dL, and alkaline phosphatase and aspartate aminotransferase ≤ 2.5 times the upper limit of normal. Patients could not have detectable antibodies to lintuzumab or active central nervous system involvement by leukemia. Patients were treated from February 2006 to March 2012 at Memorial Sloan Kettering Cancer Center on a protocol approved by the Center's institutional review board and registered with ClinicalTrial.gov (NCT00672165). All subjects gave written informed consent according to the Declaration of Helsinki.

Treatment

To limit “first pass” binding of the radiolabeled antibody in the liver, unlabeled lintuzumab was given before administration of 225Ac-lintuzumab to improve targeting of isotope to the bone marrow and other disease sites. Patients received unlabeled lintuzumab at a dose of 250 µg i.v. over 15 to 30 minutes. Fifteen to 30 minutes following infusion of unlabeled lintuzumab, 225Ac-lintuzumab was given as an i.v. infusion over 15 to 30 minutes. Five dose levels of 225Ac-lintuzumab were administered: 18.5, 37, 74, 111, or 148 kBq/kg. Initially, doses were determined based on actual body weight, but due to concerns for prolonged myelosuppression, the last six patients were treated based on adjusted body weight (ABW) if actual weight was greater than ideal body weight (IBW). IBW was calculated using standard formulas developed by Devine (18). ABW was determined using the following equation: ABW = IBW + 0.4 × (actual body weight – IBW) (19). A standard 3 + 3 dose-escalation design was used with the MTD defined as the highest dose studies for which the incidence of dose-limiting toxicity (DLT) is less than 33%. Therefore, if two or more patients in a cohort experienced DLT, the MTD was exceeded. Two to 7 patients were enrolled at each dose level. Total administered activity ranged from 888 to 14,874 kBq, and total antibody doses from 0.7 to 2.6 mg.

Given the low-level γ-emissions from 225Ac daughters 221Fr and 213Bi, radiation isolation for patients and precautions for staff were not required. Hematopoietic growth factor support was allowed according to American Society of Clinical Oncology guidelines (20). Complete blood counts and biochemical profiles were performed at least 3 times weekly. Toxicity was assessed according to the common criteria established by the NCI, version 3.0. DLTs were defined as: (i) grade 4 leukopenia lasting 35 days or more from the time of infusion (in patients with a pretreatment WBC > 1,000/μL); (ii) grade 3 or 4 thrombocytopenia that failed to recover to ≤ grade 2 at 12 weeks after treatment (in patients with a pretreatment platelet count > 50,000/μL); (iii), grade 3 elevations of alkaline phosphatase, bilirubin, aspartate aminotransferase, or alanine transaminase lasting ≥ 7 days; (iv) grade 3 elevations of serum creatinine that occurred within 6 weeks of treatment; (v) grade 2 elevations of serum creatinine lasting ≥ 7 days that occurred after 6 weeks from the time of treatment, not related to subsequent therapy; (vi) grade 3 proteinuria; (vii) other grade 3 nonhematologic toxicities lasting ≥ 10 days; or (viii) any grade 4 nonhematologic toxicity.

We evaluated pharmacokinetics of 225Ac-lintuzumab using the methods described below. To measure the antileukemic effects of 225Ac-lintuzumab, a bone marrow aspiration and biopsy was performed approximately 4 weeks after treatment. Responses were determined using the International Working Group criteria (21). Antileukemic activity was defined as elimination of peripheral blood blasts or ≥ 10% reduction of bone marrow blasts.

Pharmacokinetics

The first patient at each dose level underwent detailed pharmacokinetic studies to determine elimination kinetics in whole blood, plasma, and urine. Blood samples were collected at 5, 10, 30, 60, 90, 120, and 240 minutes after infusion of 225Ac-lintuzumab. Additional samples were obtained on days 2 and 3, day 6 or 7, and day 9 or 10 if detectable activity remained. Urine samples were obtained approximately 1 hour and 3 to 4 hours after treatment with additional samples collected on days 2 and 3, day 6 or 7, and day 9 or 10. Daughter retention and redistribution were studied by gamma counting at 2 energy windows for 221Fr (185–250 KeV) and 213Bi (360–480 KeV) (Compugamma model 1282; LKB Wallac). The data were decay-corrected to the time of injection, expressed as a percentage of the injected activity per liter, and fit to a 2-exponential decay curve.

Data availability statement

The data sets generated in this study and supporting clinical trial documents are available from the corresponding author upon reasonable request.

Patient characteristics

Eighteen patients (median age, 64 years; range, 45–80) were treated with 225Ac-lintuzumab (18.5–148 kBq/kg). Eleven patients had relapsed AML and 7 had primary refractory disease (Table 1). According to the Cancer and Leukemia Group B risk classification system, 1 patient (6%) had favorable-risk cytogenetics, 13 patients (72%) had an intermediate-risk karyotype, and 4 (22%) had poor-risk cytogenetic abnormalities (22). Molecular genetic studies were not performed. Three patients each received 18.5, 37, and 74 kBq/kg. After the MTD was exceeded in 2 patients receiving 148 kBq/kg, an additional dose level of 111 kBq/kg was studied. Among the 7 patients enrolled at this dose level, 6 patients received 111 kBq/kg. One patient enrolled at this dose level received only 45% to 50% of the intended dose due to a faulty in-line filter and was excluded from assessment for MTD determination.

Table 1.

Patient characteristics.

No. of patients (n = 18)%
Median age (range), y 64 (45–80)  
Gender 
 Male 10 56 
 Female 44 
Disease status   
 Relapsed 11 61 
  Median CR1 duration (range), months 9 (3–19)  
 Primary refractory 39 
 Median no. of prior inductions (range) 2 (1–4)  
Risk group 
 Favorable 
 Intermediate 13 72 
 Poor 22 
Median CD33 expression (range), % 91 (32–99)  
No. of patients (n = 18)%
Median age (range), y 64 (45–80)  
Gender 
 Male 10 56 
 Female 44 
Disease status   
 Relapsed 11 61 
  Median CR1 duration (range), months 9 (3–19)  
 Primary refractory 39 
 Median no. of prior inductions (range) 2 (1–4)  
Risk group 
 Favorable 
 Intermediate 13 72 
 Poor 22 
Median CD33 expression (range), % 91 (32–99)  

Adverse effects

Myelosuppression was difficult to evaluate in these patients due to coexisting baseline suppression of hematopoiesis by active AML. Only 7 patients (1 patient at 18.5 kBq/kg, 2 at 74 kBq/kg, and 4 at 111 kBq/kg) entered the study with baseline neutrophil counts of ≥ 500/µL and were considered evaluable. Six of these patients developed grade 4 neutropenia after treatment with 74 or 111 kBq/kg (Table 2). Except for 1 patient, all were red blood cell transfusion dependent at the time of study entry. This subject developed grade 3 anemia after treatment with 37 kBq/kg. Only 6 patients were platelet transfusion independent at the time of study entry (1 patient at 18.5 kBq/kg, 2 at 74 kBq/kg, and 3 at 111 kBq/kg). Grade 4 thrombocytopenia after treatment was seen in all 6 patients. The duration of neutropenia and thrombocytopenia was difficult to assess as all but 3 patients had more than 5% blasts on bone marrow aspirations following treatment. Nevertheless, the median time to resolution of grade 4 leukopenia among 13 patients who could be evaluated across all dose levels regardless of response was 27 days (range, 0–71 days). The duration of myelosuppression was unrelated to the level of CD33 expression (r = 0.067; P = 0.846) or number of prior treatments (r = 0.537; P = 0.089) but correlated with administered activity (r = 0.784; P = 0.007).

Table 2.

Serious treatment-emergent adverse effects.

Dose level (kBq/kg)
18.53774111148
Event(n = 3)(n = 3)(n = 3)(n = 7a)(n = 2)
Neutropenia (grade 4) NE NE NE 
Thrombocytopenia (grade 4) NE NE 
Myelosuppression > 35 daysb 
Febrile neutropenia (grade 3) 
Bacteremia (grade 3 or 4) 
Pneumonia (grade 3) 
Sinusitis (grade 3) 
Cellulitis (grade 3) 
Colitis (grade 3) 
Death due to sepsisb 
Alkaline phosphatase (grade 3) 
Bilirubin (grade 3) 
Atrial fibrillation/flutter (grade 3 or 4) 
Syncope (grade 3) 
CNS hemorrhage (grade 3) 
Dose level (kBq/kg)
18.53774111148
Event(n = 3)(n = 3)(n = 3)(n = 7a)(n = 2)
Neutropenia (grade 4) NE NE NE 
Thrombocytopenia (grade 4) NE NE 
Myelosuppression > 35 daysb 
Febrile neutropenia (grade 3) 
Bacteremia (grade 3 or 4) 
Pneumonia (grade 3) 
Sinusitis (grade 3) 
Cellulitis (grade 3) 
Colitis (grade 3) 
Death due to sepsisb 
Alkaline phosphatase (grade 3) 
Bilirubin (grade 3) 
Atrial fibrillation/flutter (grade 3 or 4) 
Syncope (grade 3) 
CNS hemorrhage (grade 3) 

Abbreviation: NE, not evaluable due to baseline cytopenias.

aIncludes 1 patient who received only approximately 45%–50% of the planned activity.

bDose-limiting toxicity.

As expected, neutropenic fever and infections were commonly seen (Table 2). Febrile neutropenia was seen in 6 patients (33%) at all dose levels except 74 kBq/kg. Six patients (33%) had bacteremia (1 patient after 37 kBq/kg; 4 patients after 111 kBq/kg, including 1 patient who received only a partial dose and another with a catheter-related infection; and 1 patient after 148 kBq/kg). Three patients (17%) developed grade 3 pneumonia after receiving 18.5, 74, and 111 kBq/kg. Other infections included sinusitis in 1 patient receiving 111 kBq/kg, cellulitis in 2 patients treated with 74 and 148 kBq/kg, and colitis in 1 patient after receiving 148 kBq/kg. Death due to sepsis was seen in 2 patients, 1 patient each at the 111 and 148 kBq/kg dose levels 24 and 28 days following treatment, respectively.

One patient with a history of fungal pneumonia and fungal hepatitis prior to treatment on study developed grade 3 elevation in alkaline phosphatase 32 days after receiving 37 kBq/kg of 225Ac-lintuzumab. This resolved in 6 days after discontinuing voriconazole and micafungin. Additionally, 1 patient treated with 74 kBq/kg experienced grade 2 elevation in alkaline phosphatase (occurring 19 days after treatment and lasting 7 days) and grade 2 increase in bilirubin (occurring 14 days after treatment and lasting 9 days). Grade 3 hyperbilirubinemia occurring 25 days after 225Ac-lintuzumab treatment was seen in 1 patient after receiving 111 kBq/kg, 3 days prior to death from sepsis and multiorgan failure. Grade 3 hyperbilirubinemia also occurred in 1 patient 24 days following treatment with 148 µCi/kg in the setting of sepsis and treatment with multiple antibiotics. The patient died 2 days later after antibiotic support was withdrawn. Two patients developed atrial fibrillation/flutter in the setting of infection 3 and 25 days following doses of 37 and 111 kBq/kg, respectively. Following treatment with 148 kBq/kg, 1 patient had a syncopal episode (grade 3) likely related to orthostasis, complicated by an asymptomatic subarachnoid hemorrhage (grade 3). Of note, no renal toxicity, manifested by either grade 3 or 4 elevations in serum creatinine or proteinuria, was seen with follow-up between 1 and 24 months (median, 3 months).

Based on these data, we determined that 148 kBq/kg exceeded the MTD of 225Ac-lintuzumab. DLT occurred in both patients treated at this dose level. One patient died from sepsis as noted above and a second had prolonged marrow aplasia lasting 71 days. We determined the MTD of a single infusion of 225Ac-lintuzumab to be 111 kBq/kg. Among the 6 patients receiving this dose, DLT was seen in only 1 patient. This patient developed pneumonia, sepsis, and multiorgan failure leading to death 28 days after receiving 225Ac-lintuzumab. Importantly, this morbidly obese patient received the highest administered activity of 225Ac in the study, even greater than the 2 patients treated with 148 kBq/kg. This event subsequently led to a protocol modification with dosing based on ABW for patients whose actual body weight exceeded their IBW.

Pharmacokinetics

Two-phase elimination kinetics for 225Ac-lintuzumab in whole blood, plasma, and urine were similar at all dose levels. The mean biological plasma half-lives t1/2 − α and t1/2 − β for 225Ac-lintuzumab were 1.9 and 38 hours, respectively (Fig. 2). The mean percentage clearance for the α half-life was 74.9% ± 5.77%. The mean percentage clearance for the β half-life was 25.1% ± 5.77%. These results are comparable with the results with 131I-lintuzumab (2). In the case of 213Bi-lintuzumab, the kinetics are dominated by the short physical half-life of 213Bi (46 minutes) that results in a short effective plasma half-life (12). The low administered activities of 225Ac precluded gamma camera imaging for biodistribution studies.

Figure 2.

Percentage of injected dose per liter of 225Ac-lintuzumab in whole blood, plasma, and urine over time. Data shown are for patient no. 1 following infusion of 225Ac-lintuzumab 1,110 kBq (18.5 kBq/kg). Similar pharmacokinetics were observed across all dose levels.

Figure 2.

Percentage of injected dose per liter of 225Ac-lintuzumab in whole blood, plasma, and urine over time. Data shown are for patient no. 1 following infusion of 225Ac-lintuzumab 1,110 kBq (18.5 kBq/kg). Similar pharmacokinetics were observed across all dose levels.

Close modal

Antileukemic activity

Patients underwent bone marrow aspirations and biopsies at baseline and approximately 4 weeks after administration of 225Ac-lintuzumab. Not all patients had evaluable specimens at both time points. Although remissions were not observed, antileukemic effects, defined as elimination of peripheral blood blasts or a ≥ 10% reduction in the percentage of marrow blasts, were seen across all dose levels. Peripheral blood blasts were eliminated in 10 of 16 evaluable patients (63%) but only at doses of at least 37 kBq/kg. Bone marrow blasts were reduced in 10 of 15 patients (67%) who were evaluated (Fig. 3). Eight of these patients (53%) had reductions of 50% or more. Three patients (20%), treated with 37, 111, and 148 kBq/kg, had 5% or fewer bone marrow blasts 1 month after administration of 225Ac-lintuzumab. The patient receiving 37 kBq/kg achieved a morphologic leukemia-free state. He subsequently underwent allogeneic HCT and died of transplant-related complications, including veno-occlusive disease (VOD). The patient treated with 111 kBq/kg also underwent allogeneic HCT with residual 5% blasts and subsequently died of infectious complications. The patient receiving 148 kBq/kg had marrow aplasia 1 month after therapy and recovered with leukemic blasts 2.4 months following treatment. The percentage reduction in marrow blasts did not correlate with baseline blast percentage (r = 0.392; P = 0.148), CD33 expression (r = −0.282; P = 0.308), or injected activity (r = −0.1; P = 0.738). Median overall survival for all patients was 3 months (range, 1–24 months).

Figure 3.

Percentage change in bone marrow blasts after treatment with 225Ac-lintuzumab in 15 evaluable patients.

Figure 3.

Percentage change in bone marrow blasts after treatment with 225Ac-lintuzumab in 15 evaluable patients.

Close modal

This is the first study to show that targeted therapy with an in vivo α-particle generator is feasible in patients with AML. Although no remissions were seen in this study, 225Ac-lintuzumab was shown to have antileukemic activity across all dose levels studied, manifested by elimination in peripheral blood blasts or reductions in bone marrow blasts. These results, as well as previous results with 213Bi-lintuzumab, clearly support further investigation of targeted α-particle therapy with 225Ac-lintuzumab in a variety of clinical settings either alone or in combination with standard chemotherapy and novel targeted agents.

Bone marrow blasts were reduced in 10 of 15 (67%) evaluable patients across all dose levels, 8 of whom had reductions of 50% or more. Although responses did not correlate with injected activities, 10 of 16 evaluable patients (63%) had elimination of peripheral blood blasts, but only at doses of at least 37 kBq/kg. Lack of correlation between injected activity and marrow blast reduction in this study may be explained by the small number of patients with wide variability in tumor burden, CD33 expression, and underlying radiation resistance due in part to prior therapy.

Therapy with high-energy, short-range α-particles can potentially provide for enhanced tumor cell killing and simultaneously reduce the nonspecific cytotoxic effects seen with β-emitters. Despite these advantages, it is not surprising that complete remissions after a single infusion of 225Ac-lintuzumab were not seen in this study. Assuming a leukemic tumor burden of 1011–12 cells with an average CD33 density of 10,000/cell results in approximately 1015–16 binding sites available to lintuzumab in a patient with overt AML. With about 5 DOTA per IgG and 1 in 5,000 DOTA molecules coordinating an 225Ac molecule, only 0.1% of lintuzumab molecules carried the radiolabel (17). At the administered activities and range of specific activities used in this study including 0.25 mg of cold lintuzumab, the amount of antibody infused was unable to saturate all of the available CD33 sites. Therefore, despite the potency of the isotope, at these low specific activities it was difficult to deliver enough 225Ac atoms (perhaps 3–5) to every leukemia necessary for efficient cell killing. This supports the study of 225Ac-lintuzumab at higher specific activities, in combination with other agents, or for elimination of measurable residual disease after cytoreduction.

We determined the MTD for a single infusion of 225Ac-lintuzumab to be 111 kBq/kg. Myelosuppression and infectious complications, and not extramedullary toxicity, were dose limiting. Prolonged myelosuppression (>35 days) and death due to sepsis within 30 days of treatment were seen in both patients who received a dose of 148 kBq/kg. Of concern prior to this study was the possibility of renal toxicity due to uptake of free 213Bi by the kidneys, which was seen in preclinical testing in nonhuman primates (23). No renal toxicity manifested by either grade 3 or 4 elevations in serum creatinine or proteinuria was seen with follow-up between 1 and 24 months. Because CD33 in nonhuman primates is not recognized by lintuzumab, 225Ac-lintuzumab had a significantly longer half-life in these animals. Moreover, once internalized, 225Ac and its daughters are retained by target cells. These factors account for the increased toxicity in animal studies compared with what was observed in this study.

Although only 3 patients (17%) experienced short-lived grade ≥ 3 liver function abnormalities after receiving 225Ac-lintuzumab, 1 of 2 patients who subsequently underwent HCT developed VOD. Although VOD is a known complication of transplant conditioning regimens, leukemia cells and CD33-expressing reticuloendothelial cells in the liver could potentially result in an increased risk of hepatic toxicity following 225Ac-lintuzumab. Significant hepatic toxicity has been associated with another CD33-targeting agent, gemtuzumab ozogamicin (GO), an antibody–drug conjugate containing the antitumor antibiotic calicheamicin. In initial studies, treatment with GO resulted in grade 3 or 4 hyperbilirubinemia in 29% of patients and elevated transaminases in 18%, presumably due to dissociation of calicheamicin in the liver. VOD occurred in 5% of patients, predominantly in patients who underwent HCT (24). These safety concerns ultimately led to withdrawal of GO from the market. Additional studies, however, showed the safety and benefit of the drug using lower doses in a fractionated schedule, resulting in its reapproval (25). Given the limited number of patients in this study, particularly those undergoing HCT, it is difficult to assess the risk of VOD after 225Ac-lintuzumab. Nevertheless, given the potential for hepatic toxicity and VOD associated with this agent, careful monitoring for these events is essential in future studies of 225Ac-lintuzumab.

The second-generation construct of 225Ac-lintuzumab was developed to overcome 2 major obstacles for widespread use of 213Bi-lintuzumab, including the need for an 225Ac/213Bi generator onsite and its short half-life. By directly conjugating the isotope generator to a tumor-specific antibody, 225Ac serves as an in vivo generator of 4 α-particles. The 10-day half-life and 4 α-particle emission explain the increased potency seen with 225Ac constructs compared with 213Bi analogues (14). Based on the antileukemic activity seen here and the previously demonstrated benefit to sequential therapy with cytarabine followed by 213Bi-lintuzumab, a phase I/II study of low-dose cytarabine and 225Ac-lintuzumab in older patients with untreated AML was conducted.

Like most anticancer therapies, we expect that 225Ac-lintuzmab will be most effective when integrated into rationally designed treatment programs in combination with cytotoxic chemotherapy or other targeted agents. Given the potential for myelosuppression, however, careful phase I studies will be required to optimize dose and treatment schedules in distinct clinical settings. Additionally, lower doses of 225Ac-lintuzumab as a single agent for elimination of measurable residual disease may mitigate the myelosuppression seen in the setting of overt disease, but dose-finding studies will again be needed to determine the optimal dose that can be given to individuals with normal hematopoiesis at the time of treatment. Finally, 225Ac-lintuzumab could potentially be used to intensify conditioning before HCT with the possibility of decreased extramedullary toxicity for AML MDS, and in other CD33-expressing malignances. Higher doses may be possible in this setting but careful monitoring for hepatic toxicity, including VOD, will be critical.

M.R. McDevitt reports other support from Pharmactinium during the conduct of the study; in addition, M.R. McDevitt has a patent for Actinium and the use thereof licensed and with royalties paid from Pharmactinium. J.A. Carrasquillo reports grants from Memorial Sloan Kettering Cancer Center during the conduct of the study and has been consultant to Y-mAbs, a company which has been developing antibodies and radiolabeled antibodies for therapy of cancer; those antibodies are unrelated to the antibody used in this trial or to 225Ac. N. Pandit-Taskar reports grants and other support from Bayer; grants and personal fees from Imaginab, Illumina-Innervate, and Actinium Pharma; and grants from Fusion Pharma, Clarity Pharma, and Janssen outside the submitted work. M.G. Frattini reports other support from Cellectis, Inc. and BMS outside the submitted work. P.G. Maslak reports grants from API during the conduct of the study, as well as grants from BD Biosciences outside the submitted work. J.H. Park reports personal fees from Amgen, Novartis, Kite Pharma, Pfizer, BMS, Kura Oncology, Allogene, Curocel, Artiva, Intellia Therapeutics, Autolus, Servier, Affyimmune, Minerva, and PrecisionBio outside the submitted work. S.M. Larson reports grants and other support from Y-mAbs Therapeutics Inc and Elucida Oncology outside the submitted work. In addition, S.M. Larson has a patent for MSKCC issued and licensed to Y-mAbs Therapeutics and a patent for MSKCC issued and licensed to Samus Therapeutics LLC; please see https://www.mskcc.org/research/ski/labs/steven-larson. D.A. Scheinberg reports grants and other support from Actinium during the conduct of the study, as well as personal fees from Actinium, Eureka, Pfizer, Sellas, and Repertoire outside the submitted work; in addition, D.A. Scheinberg has a patent for Alpha-particle technology issued and licensed to Actinium. J.G. Jurcic reports grants and non-financial support from Actinium Pharmaceuticals, Inc., during the conduct of the study. J.G. Jurcic also reports grants from Astellas Pharma and Forma Therapeutics, Gilead/Forty Seven, Celularity, GlycoMimetics, Arog, PTC Therapeutics, and Genetech; grants and personal fees from Syros Pharmaceuticals, AbbVie, and BMS/Celgene; and personal fees from Novartis outside the submitted work. In addition, J.G. Jurcic is a Clinical Advisory Board member for Actinium Pharmaceuticals, Inc. (without financial compensation). No disclosures were reported by the other authors.

T.L. Rosenblat: Data curation, formal analysis, investigation, writing–original draft, writing–review and editing. M.R. McDevitt: Investigation, methodology, writing–review and editing. J.A. Carrasquillo: Investigation, writing–review and editing. N. Pandit-Taskar: Investigation, writing–review and editing. M.G. Frattini: Investigation, writing–review and editing. P.G. Maslak: Investigation, writing–review and editing. J.H. Park: Supervision, investigation, writing–review and editing. D. Douer: Supervision, investigation, writing–review and editing. D. Cicic: Resources, formal analysis, funding acquisition, project administration, writing–review and editing. S.M. Larson: Formal analysis, supervision, investigation, writing–review and editing. D.A. Scheinberg: Conceptualization, resources, formal analysis, supervision, investigation, writing–review and editing. J.G. Jurcic: Conceptualization, data curation, formal analysis, supervision, investigation, methodology, writing–review and editing.

We thank Renier Brentjens, Ellin Berman, Nicole Lamanna, and Mark Weiss for clinical care; Michael Curcio, Jing Qiao, and Eva Burnazi for laboratory assistance; Suzanne M. Chanel, Amabelle Lindo, and Louise Harris for research nursing support; Kevin Zikaras for data management; Nuclear Medicine Radiopharmacists led by Rashid Ghani for drug dispensing; Saed Mirzadeh (Oak Ridge National Laboratory, Oak Ridge, TN) for supplying 225Ac; and Mark Berger (Actinium Pharmaceuticals, Inc.) for his helpful comments. This work was supported by NIH grants PO1 CA33049 and RO1 55349, the Lymphoma Foundation, and Actinium Pharmaceuticals, Inc. (New York, NY).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Howlader
N
,
Noone
AM
,
Krapcho
M
,
Miller
D
,
Brest
A
,
Yu
M
, et al
.
(eds)
.
SEER Cancer Statistics Review, 1975–2017
.
Bethesda, MD
:
NCI.
https://seer.cancer.gov/csr/1975_2017/,
based on November 2019 SEER data submission, posted to the SEER web site
,
April 2020
.
2.
Caron
PC
,
Jurcic
JG
,
Scott
AM
,
Finn
RD
,
Divgi
CR
,
Graham
MC
, et al
.
A phase 1B trial of humanized monoclonal antibody M195 (anti-CD33) in myeloid leukemia: specific targeting without immunogenicity
.
Blood
1994
;
83
:
1760
8
.
3.
Jurcic
JG
,
DeBlasio
T
,
Dumont
L
,
Yao
TJ
,
Scheinberg
DA
.
Molecular remission induction with retinoic acid and anti-CD33 monoclonal antibody HuM195 in acute promyelocytic leukemia
.
Clin Cancer Res
2000
;
6
:
372
80
.
4.
Caron
PC
,
Dumont
L
,
Scheinberg
DA
.
Supersaturating infusional humanized anti-CD33 monoclonal antibody HuM195 in myelogenous leukemia
.
Clin Cancer Res
1998
;
4
:
1421
8
.
5.
Feldman
E
,
Kalaycio
M
,
Weiner
G
,
Frankel
S
,
Schulman
P
,
Schwartzbert
L
, et al
.
Treatment of relapsed or refractory acute myeloid leukemia with humanized anti-CD33 monoclonal antibody HuM195
.
Leukemia
2003
;
17
:
314
8
.
6.
Raza
A
,
Jurcic
JG
,
Roboz
GJ
,
Maris
M
,
Stephenson
JJ
,
Wood
BL
, et al
.
Complete remissions observed in acute myeloid leukemia following prolonged exposure to lintuzumab: a phase 1 trial
.
Leuk Lymph
2009
;
50
:
1336
44
.
7.
Feldman
EJ
,
Brandwein
J
,
Stone
R
,
Kalaycio
M
,
Moore
J
, O'Connor J, et al
.
Phase III randomized multicenter study of a humanized anti-CD33 monoclonal antibody, lintuzumab, in combination with chemotherapy, versus chemotherapy alone in patients with refractory or first-relapsed acute myeloid leukemia
.
J Clin Oncol
2005
;
23
:
4110
6
.
8.
Sekeres
MA
,
Lancet
JE
,
Wood
BL
,
Grove
LE
,
Sandalic
L
,
Sievers
EL
., et al
.
Randomized phase IIb study of low-dose cytarabine and lintuzumab versus low-dose cytarabine and placebo in older adults with untreated acute myeloid leukemia
.
Haematologica
2013
;
98
:
119
28
.
9.
Burke
JM
,
Caron
PC
,
Papadopoulos
EB
,
Divgi
CR
,
Sgouros
G
,
Panageas
KS
, et al
.
Cytoreduction with iodine-131-anti-CD33 antibodies before bone marrow transplantation for advanced myeloid leukemias
.
Bone Marrow Transplant
2003
;
32
:
549
56
.
10.
Jurcic
JG
,
Divgi
CR
,
McDevitt
MR
,
Ma
D
,
Sgouros
G
,
Finn
RD
, et al
.
Potential for myeloablation with yttrium-90-HuM195 (anti-CD33) in myeloid leukemia [abstract
].
Proc Am Soc Clin Oncol
2000
;
19
:
8a
.
11.
Sgouros
G
,
Hobbs
R
,
Josefsson
A
.
Dosimetry and radiobiology of alpha-particle emitting radionuclides
.
Curr Radiopharm
2018
;
11
:
209
14
.
12.
Jurcic
JG
,
Larson
SM
,
Sgouros
G
,
McDevitt
MR
,
Finn
RD
,
Divgi
CR
, et al
.
Targeted alpha particle immunotherapy for myeloid leukemia
.
Blood
2002
;
100
:
1233
9
.
13.
Rosenblat
TL
,
McDevitt
MR
,
Mulford
DA
,
Pandit-Taskar
N
,
Divgi
CR
,
Panageas
KS
, et al
.
Sequential cytarabine and alpha-particle immunotherapy with bismuth-213-lintuzumab (HuM195) for acute myeloid leukemia
.
Clin Cancer Res
2010
;
16
:
5303
11
.
14.
McDevitt
MR
,
Ma
D
,
Lai
LT
,
Simon
J
,
Borchardt
P
,
Frank
RK
, et al
.
Tumor therapy with targeted atomic nanogenerators
.
Science
2001
;
294
:
1537
40
.
15.
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
.
16.
Sathekge
M
,
Bruchertseifer
F
,
Knoesen
O
,
Reyneke
F
,
Lawal
I
,
Lengana
T
, et al
.
225Ac-PSMA-617 in chemotherapy-naive patients with advanced prostate cancer: a pilot study
.
Eur J Nucl Med Mol Imaging
2019
;
46
:
129
38
.
17.
McDevitt
MR
,
Ma
D
,
Simon
J
,
Frank
RK
,
Scheinberg
DA
.
Design and synthesis of 225Ac radioimmunopharmaceuticals
.
Appl Radiat and Isot
2002
;
57
:
841
7
.
18.
Devine
BJ
.
Gentamicin therapy
.
Drug Intell Clin Pharm
1974
;
8
:
650
5
.
19.
Pai
MP
,
Paloucek
FP
.
The origin of the “ideal” body weight equations
.
Ann Pharmacother
2000
;
34
:
1066
9
.
20.
Ozer
H
,
Armitage
JO
,
Bennett
CL
,
Crawford J Demetri
GD
,
Pizzo
PA
, et al
.
2000 Update of recommendations for the use of hematopoietic colony-stimulating factors: evidence-based, clinical practice guidelines
.
J Clin Oncol
2000
;
18
:
3558
85
.
21.
Cheson
BD
,
Bennett
JM
,
Kopecky
KJ
,
Büchner
T
,
Willman
CL
,
Estey
EH
, et al
.
Revised recommendations of the International Working Group for diagnosis, standardization of response criteria, treatment outcomes, and reporting standards for therapeutic trials in acute myeloid leukemia
.
J Clin Oncol
2003
;
21
:
4642
9
.
22.
Byrd
JC
,
Mrozek
K
,
Dodge
RK
,
Carroll
AJ
,
Edwards
CG
,
Arthur
DC
, et al
.
Pretreatment cytogenetic abnormalities are predictive of induction success, cumulative incidence of relapse, and overall survival in adult patients with de novo acute myeloid leukemia: results from Cancer and Leukemia Group B (CALGB 8461)
.
Blood
2002
;
100
:
4325
36
.
23.
Miederer
M
,
McDevitt
MR
,
Sgouros
G
,
Kramer
K
,
Cheung
N-KV
,
Scheinberg
DA
.
Pharmacokinetics, dosimetry and toxicity of targetable atomic generator, 225Ac-HuM195, in nonhuman primates
.
J Nucl Med
2004
;
45
:
129
37
.
24.
Bross
PF
,
Beitz
J
,
Chen
G
,
Chen
XH
,
Duffy
E
,
Kieffer
L
, et al
.
Approval summary: gemtuzumab ozogamicin in relapsed acute myeloid leukemia
.
Clin Cancer Res
2001
;
7
:
1490
6
.
25.
Castaigne
S
,
Pautas
C
,
Terré
C
,
Raffoux
E
,
Bordessoule
D
,
Bastie
JN
, et al
.
Effect of gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid leukaemia (ALFA-0701): a randomised, open-label, phase 3 study
.
Lancet
2012
;
379
:
1508
16
.