Abstract
The morbidity associated with pediatric medulloblastoma, particularly in patients who develop leptomeningeal metastases, remains high in the absence of effective therapies. Administration of substances directly into the cerebrospinal fluid (CSF) is one approach to circumvent the blood brain barrier and focus delivery of drugs to the site of tumor. However, high rates of CSF turnover prevent adequate drug accumulation and lead to rapid systemic clearance and toxicity. Here, we show that PLA-HPG nanoparticles, made with a single-emulsion, solvent evaporation process, can encapsulate talazoparib, a PARP inhibitor (BMN-673). These degradable polymer nanoparticles improve the therapeutic index when delivered intrathecally and lead to sustained drug retention in the tumor as measured with PET imaging and fluorescence microscopy. We demonstrate that administration of these particles into the CSF, alone or in combination with systemically administered temozolomide, is a highly effective therapy for tumor regression and prevention of leptomeningeal spread in xenograft mouse models of medulloblastoma. These results provide a rationale for harnessing nanoparticles for the delivery of drugs limited by brain penetration and therapeutic index and demonstrate important advantages in tolerability and efficacy for encapsulated drugs delivered locoregionally.
One Sentence Summary:
Polymer nanoparticles exhibit long circulation and excellent retention in the CSF after intrathecal administration and, when loaded with a PARP inhibitor, talazoparib, show therapeutic efficacy in preclinical models of medulloblastoma.
INTRODUCTION
Pediatric medulloblastoma (MB) tumors are devastating lesions that arise in the cerebellum or fourth ventricle of the brain and are characterized by a high rate of leptomeningeal reoccurrence (1, 2). The standard treatment regimen for MB is maximal safe tumor resection followed by craniospinal irradiation to prevent the high risk of spread across the neuroaxis. Despite advances in survival rates over the last 50 years, this treatment modality has serious adverse effects, including neurocognitive sequalae, and 30% of patients still relapse through metastatic dissemination to the leptomeninges (3). Leptomeningeal spread (LMS) remains the major cause of mortality in children with MB (4, 5).
Systemic therapies have largely been ineffective against LMS, due to poor blood brain barrier (BBB) penetration of most agents. Given that most MB recurrences develop directly adjacent to cerebrospinal fluid (CSF), there is a strong rationale for local intra-CSF drug administration by direct intrathecal (IT) or intraventricular (ICV) delivery. However, previous efforts to deliver small molecule drugs to the CSF have been limited by the rapid efflux of therapeutics from the CSF, which occurs within hours (6–8). With increased interest in the potential of site-directed delivery of nanomedicine, recent studies have used biodegradable polymer nanoparticles (NPs) for delivery of agents to the brain, often by local administration directly into the brain tissue (9–11). But many NPs are rapidly cleared from the mouse brain through the glymphatic system, with a resulting clearance half-life of only a few hours, underscoring the difficulties in designing a high-retention, long-lasting NP system (12, 13). Moreover, no studies have been conducted to date to investigate the clearance half-life and biodistribution of IT-delivered NPs in the context of MB.
Here, we have designed NPs that slowly release members of a promising new class of agents, poly(ADP-ribose) polymerase inhibitors (PARPi). Since the discovery of the role of PARP1 in repair of single-stranded DNA breaks, four PARPi have been approved for the treatment of solid tumors (14). Increased PARP1 expression has also been observed in MB biopsies and is associated with poor prognosis (15). This suggests that a PARPi may be able to overcome high interpatient tumor heterogeneity between subtypes, as well as the high intrapatient heterogeneity between primary tumor and metastases (16). Despite this potential, a PARPi has not yet been approved for CNS tumors because they do not penetrate the brain at concentrations that spare peripheral toxicity. In previous work, we demonstrated local retention of poly(lactic acid) (PLA) NPs with a grafted hyperbranched polyglycerol (HPG) corona after intra-parenchymal delivery in a variety of tissue compartments, particularly after converting the NP coronas to a bioadhesive state (17–19). This experience led us to hypothesize that we could adopt the PLA-HPG NPs to address limitations in treatment of LMS from MB. We hypothesized that by administering PLA-HPG NPs encapsulating talazoparib (BMN-673), a highly potent PARPi, directly into the CSF space, we could achieve long-retention of NPs, slow release of BMN-673, and improve tumor treatment without substantial toxicity. To test this hypothesis, we measured the biodistribution of PLA-HPG NPs in mice by two methods: positron emission tomography (PET) imaging with radiolabeled NPs and fluorescence microscopy. We discovered that NPs can be made small enough to circulate throughout the leptomeningeal space and penetrate perivascular spaces while achieving long-term retention, owing in part to NP association with the meningeal and tumor surfaces. In addition, we showed that PLA-HPG NPs loaded with BMN-673 have advantageous pharmacokinetic properties, including decreased systemic exposure to BMN-673 and selective tumor exposure, which result in an improved safety profile, increased antitumor activity, and inhibition of LMS growth in a MB xenograft mouse model.
RESULTS
Engineering NPs for optimal CNS retention
To produce PLA-HPG NPs, we used a single emulsion solvent evaporation technique that has been previously described (17); here, we modified the technique to allow us to monitor their location in vivo by fluorescence microscopy. Particle size and surface charge was determined for a range of particle compositions (Fig. 1, A–C). To synthesize covalently tethered fluorescent dye NPs for effective tracking by microscopy, NPs were produced with blends of PLA-HPG copolymer and a 10% Cy5-PLA conjugate. Unlike dyes, which may leak from NPs and complicate interpretation of measurements, Cy5-PLA conjugation ensures that fluorescent signal originates from the NPs themselves (20). We hypothesized that PLA-HPG NPs would similarly exhibit clear advantages in the CSF environment after direct CSF injection through the cisterna magna (CM). We administered Cy5-PLA-HPG NPs (Cy5-NPs) into the CM of healthy mice and prepared frozen brain and spinal cord sections 24 h and 48 h post-injection. At 48 h post injection, we detected substantial accumulation of Cy5-NPs in the leptomeninges and perivascular spaces, without parenchymal uptake in all coronal sections of the brain. We also observed an even deposition of Cy5-NPs surrounding the outer layer of the spinal cord (Fig. 1, D–E).
Figure 1 |. NP characteristics, distribution in the brain and spinal cord.

(A) Nanoparticle platform consists of a poly(lactic acid) (PLA) core surrounded by a hyperbranched polyglycerol (HPG) shell. PDLLA-Cy5 (represented by blue stars in the diagram) can be incorporated in the core for fluorophore imaging. The PLA core can be loaded with BMN-673 drug (represented by purple ovals). The HPG shell can be modified to an aldehyde-rich surface (represented by purple circles) or functionalized with DFO-mesylate (represented by red circles) for 89Zr labeling. (B) Mean dynamic light scattering measurements with standard deviation (s.d.) of hydrodynamic diameters and polydispersity index (PDI) of NPs in deionized (DI) water (n=3). (C) Mean zeta potential of NPs with s.d. (n=3). (D) Representative coronal head sections of mice showing DAPI and localization of Cy5-NPs in the olfactory epithelium, the forebrain, and the pontocerebellar region (from top to bottom), 48 h after intrathecal (IT) administration (n=5), with scale bar = 500 μm (left column) and 50 μm (right column). (E) Representative spinal cord sections of the cervical, thoracic, and lumbar regions 48 h after IT administration (n=5), with scale bar as 500 μm (left column) and 50 μm (right column). (F) J:Nu mice (n=6) were administered Cy5-NPs by a CM implanted catheter. Fluorescence images of the whole body were taken 3 h and 48 h after administration. (G) In vivo imaging system IVIS) images of Cy5-NPs and aldehyde Cy5-NPs in ex vivo organs at 24 h, 5 d, and 7 d post administration by a CM implanted catheter in J:Nu mice, shown with radiant efficiency (p/sec/cm2/sr/(PW/cm2)). (H) Comparison of the Cy5 cranial radiance intensity (p/sec/cm2/sr) as measured by IVIS between the two NP formulations at 3 h, 24 h, 5 d and 7 d., same mice as (F) and (G). No statistically significant difference in signal intensity at any time point as measured by one-way ANOVA.
Prior reports suggest that the diameter of the administered vehicle is an important element affecting CSF half-life (21, 22), so we assessed CNS retention of three distinct sizes of PLA-HPG NPs: 90 nm, 150 nm, and 210 nm (Table S1). Six hours after intra-CSF delivery by injection into the CM, mouse brain and spinal cord sections were imaged. FITC-conjugated dextran (either MW= 3- or 3,000 kDa) served as size-fractionated controls (diameter estimated to be <4 nm and >60 nm respectively) (23). Although all mice administered Cy5-NPs showed retention in the brain and spinal cord leptomeninges, the 90 nm NPs covered the greatest percentage of area in fresh frozen coronal brain sections as quantified by fluorescent signal (P < 0.005 when compared to 150 nm, P <0.0001 when compared to 210 nm) (Fig. S1). In comparison, although a strong fluorescent signal was detected for both dextrans at 30 min after injection, no signal was detected in any brain or spinal cord sections of mice that were administered the 3 kDa and 3,000 kDa dextran at 6 hours post-injection (Fig. S2).
Next, we compared the effect of surface charge and surface chemistry of the PLA-HPG NPs. Previously, we reported a method for converting the HPG on the NP surface into an aldehyderich corona with enhanced bioadhesive properties (termed aldehyde-NPs) (17, 24, 25). Previous studies showed that the conversion from NP to aldehyde-NP improved plasma half-life and organ accumulation after intravenous (i.v.) or intraperitoneal (i.p.) administration (18, 24), and an impact on local retention after topical or subcutaneous administration (25–27); this conversion had a negligible effect on CSF distribution in the leptomeningeal region (Fig. 1, F–G). There were no significant differences in CNS retention between Cy5 conjugates of the aldehyde-NPs and NPs over 7 days of ex vivo whole-body imaging with the Xenogen in vivo imaging system (IVIS). Both formulations of NPs were well retained in tumor-free brain and spinal cord of healthy mice (BALB/C) at all timepoints examined with no significant difference in fluorescent radiance flux as measured in vivo and ex vivo (Fig. 1, H). In case there could be differences in accumulation along lymphatic clearance pathways, we assessed the percentage of Cy5-NP positive cells and percentage of Cy5-NP-aldehyde positive cells in the mandibular and deep cervical lymph nodes (mLN and dCLN respectively) of tumor-free mice (BALB/C) and tumor-bearing mice (human MB cell xenografts into J:Nu homozygous Foxn1/F mice). Again, there were no significant differences in the accumulation of NPs in immune cells (CD45+). We looked closer at mononuclear phagocytes, specifically macrophages (CD11b+ CD11c−) and dendritic cells (CD11b− CD11c+ and CD11b+ CD11c+) of the lymph nodes. When considering the effect of surface chemistry on uptake by comparing aldehyde-NPs and NPs, there were no observable differences in NP accumulation within these cell populations; however, we did note more accumulation of the aldehyde-NPs with statistical significance in both dendritic cell subpopulations in mLN in WT mice (P <0.05 for CD11b− CD11c+, P <0.01 for CD11b+CD11c+) and in CD11b+ CD11c+ subpopulation in tumor-bearing mice (P <0.05) (Fig. S3).
NPs with the most optimal characteristics for CNS retention exhibited a spherical morphology under electron microscope analysis (Fig. S4), with an average hydrodynamic diameter of 90–100 nm (Fig. 1, B). This is consistent with earlier findings on NP delivery, which indicates that smaller particles are less readily cleared by NP-macrophage interactions (28–30). As such, we opted to use unmodified NPs for drug encapsulation; their zeta potential averaged around −10 mV (Fig. 1, A–C).
NPs show higher CNS retention compared to small molecules
As microscopy studies suggest that PLA-HPG NPs have potential as carriers for the delivery of drugs by IT administration, we sought to clarify the whole-body biodistribution kinetics of the NPs in the CSF space and in vital organs. To image our NPs with PET, we developed a radiolabeled NP PET probe with analogous properties. We chose positron-emitting zirconium-89 (89Zr) to label PLA-HPG NPs, due to its stability and long half-life (t1/2 = 78.4 h), and availability of a well-characterized chelator, deferoxamine (DFO) for 89Zr (31). We first functionalized the NP surface with DFO-mesylate through a Schiff-base reaction, and detected no change in hydrodynamic diameter, but did observe a small change in zeta-potential from −10 mV to −5 mV (Fig. 1, C). The DFO-conjugated NPs were subsequently used to chelate 89Zr (Fig. S5).
We measured the quantitative biodistribution of our [89Zr]Zr-DFO-NPs into all major organs over time by PET imaging in mice. We first performed a 2 h dynamic scan, to determine the distribution and timing of NP after IT administration, in BALB/C mice without tumors, using free [89Zr]Zr-DFO as a control. Within 5 min of injection, indicated as the 0 h time point, we observed NPs distributing from the site of injection (CM) into the brain and cervical regions of the spinal cord (Fig. 2, A–B). The amount of [89Zr]Zr-DFO-NP recorded in the CNS dropped slightly and then remained constant for the next 2 h, with limited distribution of signal to the systemic circulation or other organs. In contrast, [89Zr]Zr-DFO distributed immediately throughout the subarachnoid space, similar to the NPs, but then distributed from the CNS into systemic circulation, with less than 30% ID/g detectable in the CNS by 2 h post injection of the tracer. We determined that the [89Zr]Zr-DFO had a CNS half-life of ~60 min, which is comparable to known half-lives of small molecules following IT delivery (32, 33). In comparison, the amount of [89Zr]Zr-DFO-NPs were relatively stable in the brain and spinal cord during the first 2 h of injection. Additionally, [89Zr]Zr-DFO enters the interstitial parenchyma to a greater extent than [89Zr]Zr-DFO-NPs (Fig. 2, C). We think this is related to the size difference between the two materials (100 nm vs < 1 nm). We note that we observed an accumulation of [89Zr]Zr-DFO-NP in the bladder as early at 5 min post injection (Fig. 2, D), which is addressed in Supplementary Material (Fig. S6).
Figure 2 |. PET/CT of [89Zr]Zr-DFO-NPs and [89Zr]Zr-DFO delivered i.c.m. in tumor-free mice:

(A-B) Healthy BALB/C mice were injected by IT administration with either [89Zr]Zr-DFO-NP or [89Zr]Zr-DFO and imaged continuously for 2 hours (h) (n=2); (A) Pharmacokinetic curve in the central nervous system of [89Zr]Zr-DFO-NP and [89Zr]Zr-DFO, *P=0.0243; (B) Pharmacokinetic curve in the brain and spinal cord separately of [89Zr]Zr-DFO-NPs and [89Zr]Zr-DFO, ****P <0.0001 (brain), ns (spinal cord). ns = not significant, P > 0.05. (C) Whole-body sagittal plane PET images of continuous dynamic scan over 2 h for [89Zr]Zr-DFO and [89Zr]Zr-DFO-NPs. (D) Biodistribution at various timepoints from 3 h to 12 days (d) after [89Zr]Zr-DFO-NPs injection into healthy mice (n=4 for 4 d, 7 d, 12 d, and n=8 for 3 h, 24 h). Percent total is calculated as activity in region-of-interest over total injected activity. E, Top row: representative fused PET/CT images of mice at maximum intensity projection at 3 h, 24 h, 4 d, 7 d, 12 d after [89Zr]Zr-DFO-NP injection through CM. Bottom row: whole-body sagittal plane PET images of CNS only, equally scaled according to the color bar. (A,B,D), Data shown as the mean +/− s.d.;.
Next, we administered [89Zr]Zr-DFO-NPs (7.4 × 1010 Bq/g) IT to animals without tumors (BALB/C mice) and performed static PET/CT imaging at predetermined time points up to 12 days post injection (n=4) (Fig. 2, D–E). At 3 h, we found that at least 60% of the signal was in the brain, and at least 20% of the signal was in the spinal cord (>80% in the CNS). Activity in the brain gradually decreased over the first 4 d, eventually reaching 30%. This degree of activity was observed in the brain for at least 12 d, our last day of imaging. In the spinal region, the decline in activity was slower over the first 4 d, with similar activity observed at these time points, about 25%. On day 7 and 12, there was a decrease in signal to ~20% and ~10%. Ex vivo gamma counting after 12 d confirmed the in vivo imaging findings, with the brain exhibiting the highest Bq/g signal (44% of total injected dose per gram of tissue, or ID/g), then the liver (16%), spleen (34%), and cervical lymph nodes (3%) (Fig. S7).
NPs show higher CNS retention in xenograft tumor models
We next analyzed the CNS retention of [89Zr]Zr-DFO-NPs in tumor-bearing mice (J:Nu homozygous Foxn1/F). We developed a mouse model with leptomeningeal metastatic MB by IT injection of DAOY cells stably expressing luciferase into the CM. By day 14, IVIS showed tumor growth throughout the CNS, including widely disseminated tumors in both the cerebellum and spinal cord (Fig. S8, A). To establish that the presence of tumors did not result in BBB breakdown, we injected mice with Gd-DTPA MR contrast agent intravenously and imaged with MRI to confirm no BBB penetration occurred as compared to control (Fig. S8, B). Two mice were injected intravenously with Gd-DTPA and imaged by MRI after 20 minutes. There was no detectable Gd-DTPA signal in the brain, confirming an intact BBB. However, we observed acute hydrocephalus in vivo in both mice, most likely due to tumor obstruction of CSF flow. H&E images of the brain ex vivo revealed a high tumor burden in the brain cerebellum, ventricles, and in the spinal leptomeninges (Fig. S8, C–D).
We evaluated the biodistribution and uptake of [89Zr]Zr-DFO-NPs in tumors and normal tissue throughout a 21-day period using PET/CT imaging in two cohorts of mice (n=6 total) (Fig. 3, A–G). The first cohort (n=2) was scanned continuously for 120 min after administration. Within the first 5 min, 30% of the total injected dose was detected in the tumor, which gradually increased to 40% over the next 2 h. The next cohort (n=4) were scanned at predefined intervals (6 h, 24 h, 4 d, 7 d, 12 d, 21 d). We saw greater uptake in the tumor than in any other normal tissue, with 50% of total injected dose identified in the tumor site within the cerebellum (roughly 3.5 times higher % ID/g compared to the brain at day 21). Over the next 21 days, uptake in the brain and spinal cord remained stable at roughly 15% and 10% of injected dose, respectively. We observed greater accumulation in peripheral organs with the tumor bearing mice compared to healthy mice, specifically seeing accumulation in the cervical lymph nodes, peripheral lymph nodes, liver, and spleen. At no point did any of these peripheral organ accumulation exceed 20% of overall activity. In general, tumor bearing mice showed longer retention of NPs in the CNS, most likely due to uptake of NPs by tumor cells. At day 21, 40% of total ID/g remained detectable in the tumor site, which could be visualized using maximum intensity projections of the PET/CT image, which is a summed representation of radioactivity in the whole body (Fig. 3, G).
Figure 3 |. [89Zr]Zr-DFO-NPs accumulate preferentially in tumors.

(A,B), J:Nu mice inoculated with DAOY cells were injected by IT with [89Zr]Zr-DFO-NPs 3 weeks after inoculation and imaged continuously for 2 h (n=2). (A) Pharmacokinetic curve of [89Zr]Zr-DFO-NPs in the brain, spinal cord, and tumor site of tumor-bearing mice. (B) Pharmacokinetic curve of mouse cervical lymph nodes, bladder, and liver. (C) Biodistribution at various timepoints from 6 hours (h) to 21 days (d) after [89Zr]Zr-DFO-NPs in tumor-bearing mice (n=4). Percent total is calculated as activity in ROI over total injected activity. (D) Top row: representative fused PET/CT images of maximum intensity projection (MIP) at 6 h, 24 h, 4 d, 7 d, 12 d, 21 d after [89Zr]Zr-DFO-NP injection through CM. Bottom row: whole-body sagittal plane PET images, equally contrasted according to arrow bar. (E) in vivo fluorescent images (p/sec/cm2/sr) of luciferase-labeled tumor cells inoculated in mice 10 days prior to [89Zr]Zr-DFO-NP administration and PET/CT imaging. (F) Quantified bioluminescence intensity (p/sec/cm2/sr) of brain and spinal cord tumor signal from mice in E. (G) PET/CT MIP and sagittal plane PET image at day 21 with a 10X lower SUV intensity scale for visualization purposes. (H) H&E stains of cerebellum tumor and corresponding DAPI (blue)-stained images of Cy5-NPs (red) at 10X and 40X magnification. Scale bars = 500 μM (two left columns) and 25 μM (right column). (I) Quantification of Cy5-NP area fraction in 10 representative sections of whole brain of mice with or without tumors 24 hours after administration (n=5), *** P <0.001, one-way ANOVA.
We also identified NP accumulation at the cellular level by IT administration of Cy5-NPs in tumor-bearing mice and observing brain sections by microscopy (Fig. 3, H). We detected dense Cy5-NP accumulation and uptake in tumor foci at 24 h after injection; NP retention in this area of the cerebellum was not observed in healthy mice (Fig. 3, I). We also saw a lower density of NP accumulation in the leptomeninges of the brain and spinal cord of tumor-bearing mice than in healthy animals, implying that the NPs circulate through the perivascular space in a similar manner, but accumulate at significantly higher density in sites with tumor foci than in healthy brain or spinal tissue (P <0.001).
The presence of Cy5-NPs in areas of parenchymal tumor, and the accumulation of NPs in the deep and superficial cervical LNs as detected by PET raised the possibility of several transport pathways of the NPs into the tumor microenvironment and to the cervical LNs. We first examined whether NP accumulation at tumor sites was aided by the presence of immune cells. We injected Cy5-NPs into mice with tumors, and after 48 h, collected and sectioned the head before staining for F4/80 (a marker for macrophages) and Iba1 (a marker for microglia). At 48 h, we could detect the presence of activated microglia (red) and tumor associated macrophages (green) within the tumor bulk (Fig. 4, A–B) and detected colocalization of NPs with both cell types. In WT mice without tumors, we saw no significant NP accumulation in the brain parenchyma, and only in CSF-bathed regions such as the choroid plexus, without any microglia or macrophage association (Fig. 4, C). These results suggest that NP penetration and transport into the tumor bulk is aided by tumor-associated immune cells. We also observed a portion of NPs in the tumor microenvironment that were not associated with either macrophages or microglia.
Figure 4 |. Cy5-NPs colocalize with resident macrophages and infiltrating macrophages in the tumor, and colocalize with meningeal lymphatics, blood vessels, and immune cells in the meninges.

(A-B) Cy5-labeled NPs (blue) colocalize with Iba1 stained macrophages (red) and F4/80-stained macrophages (green). F4/80-stained macrophages and Cy5-labeled NPs are observed predominantly in the tumor microenvironment (unlabeled), whereas Iba1 stained macrophages are observed throughout the brain parenchyma. Representative images from a single experiment consisting of four biological replicates per group are displayed in (A) and (B), with (B) providing a more detailed view of the same region. (A) Scale bars = 100 μm. (B) scale bar = 50 μm. (C) Cy5-labeled NPs (blue) line the choroid plexus in the 3rd ventricle, with no colocalization with F4/80-stained macrophages and Iba1 stained resident macrophages in the brain parenchyma. Scale bar = 200 μm. (D-E) Representative images of the whole meninges (D), and close-up of mice (E) 48 h after administration of Cy5-NPs (blue) co-labeled with LYVE-1 (red) and CD31 (green) (n=4). (D) Scale bars = 100 mm. (E) Scale bar = 100 μm. (F-G) Representative images of the transverse sinus (F), and close-up (G) 48 h after administration of Cy5-NPs (blue) co-labeled with LYVE-1 (red) and CD45 (green). (F) Scale bar = 1 mm. (G) Scale bar = 100 μm.
We sought to examine the drainage pathway by which NPs are cleared from the CNS. To accomplish this, the meninges of the tumor-bearing mice were isolated at 48 h after i.c.m. injection of Cy5-NPs, and stained with the endothelial cell marker CD31, lymphatic vessel marker LYVE-1, and immune cell marker CD45. NPs accumulated throughout the entire meninges and particularly along the venous sinuses, especially the transverse sinus (Fig. 4, D). Along the transverse venous sinuses, NPs co-localized with lymphatic vessels (LYVE-1+CD31+) although other clusters of NPs were found not associated with meningeal lymphatic vessels or blood vessels (Fig. 4, E). We observed that NP accumulation frequently co-localized with macrophages (LYVE-1+ CD45+)(Fig. 4, F–G).
Polymeric encapsulation of BMN-673 alters the toxicity profile of drug in animal studies
As a proof-of-concept, we tested whether we could improve the pharmacokinetic (PK) properties and activity of PARPi. As our model drug, we chose BMN-673 for its potent PARP trapping properties and toxicity at very low doses. BMN-673-loading in NPs to obtain BMN-673encapsulated NPs ((BMN)NPs) varied from 1% to 5% (w/w) depending on the solvent ratios and drug to polymer ratios used during NP preparation (Fig. S9, A–B). We selected the NPs without the aldehyde-modified surface due to their similar persistence in the brain (Fig. 1, G–H). The BMN-673 release rate from these NPs was similar in both artificial CSF (aCSF) and PBS at 37 °C, averaging 60% release over 3 days (Fig. S9, C–D) in a bi-phasic manner, similar to the kinetics identified in prior studies (34–36). The relative cytotoxic activity of free BMN-673 and (BMN)NPs were determined on 3 MB cell lines: DAOY, D341, and D283. Although both agents were cytotoxic at a range of 10 nM to 1 μM, the NPs were more potent with a lower IC50 value than that of free BMN-673 (Fig. S10).
To characterize the safety of (BMN)NPs in vivo, we conducted toxicology studies in healthy female nude mice. We first determined the maximum tolerated dose (MTD) for single IT dosing of (BMN)NPs or free BMN-673 in nude mice (Fig. 5, A–B). Increasing doses of either (BMN)NP or free BMN-673 were administered to animals; body weight loss and overall animal health were closely monitored. MTD values for (BMN)NPs were tenfold higher than that of free BMN-673. The median lethal dose for IT administration of free BMN-673 was 0.125 mg/kg. Animals treated with greater than 0.06 mg/kg developed a variety of toxicity-related symptoms, including lethargy, labored breathing and, in some cases, death. The MTD (with a single dose) was determined to be 0.05 mg/kg. A slightly lower dose, 0.03 mg/kg twice a week, was tolerated with less than 10% body weight loss. By comparison, (BMN)NPs were well tolerated at all doses tested at or under 0.5 mg/kg, which was the maximum dose allowed in one infusion due to IT volume dosing limits. To achieve IT doses of more than 0.5 mg/kg, the mice were dosed multiple times in the same day, within 3 h, and the lethal dose of 1.25 mg/kg was determined. At 1.25 mg/kg, we noticed a delay in the onset of acute toxicity symptoms, which was presumably due to delayed drug release from the NPs.
Figure 5 |. Toxicity of BMN-673 free drug and (BMN)NPs in mice.

(A-B) Tolerability of various doses of free BMN-673 and (BMN)NPs in non-tumor bearing J:Nu mice was assessed by monitoring body weight and overall health conditions after a single IT treatment. The dose was administered to one mouse first, and if tolerated, was expanded to n=5. Black asterisk indicates animals that died or were sacrificed due to excessive weight loss. (A) Free BMN-673 was lethal at any dose at or higher than 0.06 mg/kg. Animals experienced weight loss less than 15% at 0.05 mg/kg. (B) (BMN)NPs was lethal at 1.25 mg/kg, but at lower doses weight loss was not greater than 10%. (C) J:Nu mice were treated with BMN-673 or (BMN)NPs at the MTD, and complete blood cell counts, differential white blood cell counts, and platelet cell counts were performed to evaluate hematological toxicity. For this study, n=6 for each group.
To further test systemic toxicity differences between (BMN)NPs and free BMN-673, we monitored complete blood count parameters, which measure red blood cell (RBC), white blood cell (WBC), and platelet (PLT), in mice at day 3 and day 7 following two bi-weekly administrations at MTD (Fig. 5, C). Mice treated with free BMN-673 at 0.05 mg/kg/week had progressive leukopenia and thrombocytopenia at day 3 which did not improve appreciably at day 7. NPs dosed tenfold higher, at 0.5 mg/kg/week, maintained considerably more normal WBC, RBC, and PLT counts over the course of treatment, and all cell counts except for eosinophils returned to normal ranges by day 7.
BMN-673-encapsulated NPs show superior activity compared to free BMN-673 in xenograft tumor model
We sought to translate the improved therapeutic index of (BMN)NPs over free BMN-673 to improve effectiveness in J:Nu tumor xenograft models. For in vivo studies, we performed intra-cisternal transplantation of DAOY cells that stably express luciferase. A surgical catheter was implanted into their CM and used for both cellular implantation and IT dosage during treatment. Mice were treated when their tumoral luminescence burden was detectable at 105 BLI (p/sec/cm2/sr), 7 days post-implantation. Mice were treated once, at the same dose of 0.1 mg/kg with either (BMN)NPs or free BMN-673 (Fig. 6A). (BMN)NP-treated tumors grew at a slower rate than free BMN-673-treated tumors. Tumor BLI signal was most reduced at a week after the treatment, which delayed tumor growth in subsequent weeks in contrast to the free BMN-673 group (Fig. 6B and C). Even though the dose was more than the MTD for free BMN-673, a tumor reduction benefit was observed in only 20% of mice. Consistent with the IVIS findings, mice treated with (BMN)NPs lived significantly longer than those treated with free drug alone (P <0.0001), with enhanced median survival of 56 days (Fig. 6D). Furthermore, the free BMN-673-treated mice lost more weight than the (BMN)NP group (around 20% vs. non-notable) (Fig. 6E).
Figure 6 |. Tumor inhibition of BMN-673 and (BMN)NPs with single dose in MB mouse xenograft.

(A) Timeline for the inoculation and treatment of DAOY tumor models. J:nu mice were implanted with tumor through a CM catheter, then treated using the same catheter with either free BMN-673 (0.05 mg/kg at one dose) or (BMN)NPs (0.25 mg/kg at one dose). Two mice were removed from overall study due to no observable tumor growth. Log-rank test, followed by repeated-measures ANOVA *** P <0.001. (B) Region-of-interest analysis of bioluminescence intensities (p/sec/cm2/sr) from whole brain of the course of 4 weeks. (C) Whole body bioluminescence images of DAOY tumor bearing mice taken at the end of each week. Bioluminescence scale (p/sec/cm2/sr) for 1st row is different from all remaining images. (D) Survival curves for BMN-673 treated, (BMN)NP treated, and control mice (n=6 per group, one mouse removed from control and BMN free group for no tumor growth). (E) Change in body weights of all groups. Data presented as mean +/− s.d.
We repeated this study using the same mouse model, but with twice-weekly dosing of 0.25 mg/kg of (BMN)NPs or 0.03 mg/kg for free BMN-673 (Fig. 7A), which is the amount of drug encapsulated in the BMN(NP) dosage. These equitoxic doses are equivalent to roughly 50% of MTD, and we observed similar extents of mean weight loss in both treatment groups (such as 3% at week 1 and 5% at week 2, Fig. 7B). In the NP-treated group, we observed tumor regression. The NP treatment had a substantial antitumoral impact in all the mice, especially one week after the doses were administered (Fig. 7C). (BMN)NPs led to no detectable tumor burden in several treated animals. In contrast, tumors continued to progress with treatment of free drug. In NP-treated mice where the tumor progressed, we still observed a median survival benefit of 5 weeks compared to the free drug-treated group, and a survival benefit of 6 weeks compared to the untreated control group (Fig. 7A). In this study, median survival in all treatment groups generally reflected the tumor growth rates evaluated by bioluminescence, suggesting that the mice succumbed to cancer rather than drug-induced side effects. In addition,(BMN)NPs, and to a lesser degree, free BMN-673 (P <0.05), significantly reduced the incidence of CNS metastases (P <0.0001) (Fig. 7D). We observed this improvement in rates of distant metastatic dissemination in the leptomeninges when measured by IVIS as well (Fig. 7E). In comparison, over 80% of untreated animals developed evident spinal cord dissemination and required euthanasia around week 4, due to severe hydrocephalus.
Figure 7 |. Tumor inhibition and anti-metastatic capacity of (BMN)NPs with repeat dosing in the MB mouse xenograft.

(A) Survival curves for BMN-673 treated (n=7), (BMN)NP treated (n=8), and control J:Nu mice (n = 9). Log-rank test, followed by repeated-measures ANOVA **** P <0.0001 (B) Change in body weights of all groups. Data presented as mean +/− s.d. (C) Region-of-interest analysis of bioluminescence intensities (p/sec/cm2/sr) from whole brain. (D) Percentage of mice with observable leptomeningeal spread through IVIS by 14 days post inoculation (n=16 for control, n=10 for NP and free). **** P <0.0001, *P < 0.05, one-way ANOVA. Bioluminesence images can be found in Figure S7, (A. E) Whole body bioluminescence images of DAOY tumor bearing mice. Bioluminescence scale is different for first 2 rows versus last 2 rows.
BMN-673-encapsulated NPs synergize with Temozolomide when given in conjunction in a xenograft model
To demonstrate sensitivity of the MB cell lines to TMZ and BMN-673 synergy, we first treated DAOY, D341, and D283 cells with free BMN-673 and (BMN)NPs and performed conventional IC50 analyses. When we calculated combinatorial indices using these two drugs, cells treated with incremental BMN-673 increased in the presence of TMZ demonstrated reduced viability, as quantified with the CellTiter-Glo viability assay, compared to cells treated with BMN-673 alone. When we calculated BMN-673 and TMZ combinatorial index values using a classical Loewe synergy model, we detected high synergy. Drug interaction assays with other commonly used, brain penetrant chemotherapy agents, such as Lomustine, Topotecan, Irinotecan, Cyclophosphamide, did not show consistent synergistic action with BMN-673 across all cell lines (Fig. S11).
We verified the synergy observed in vitro using a D341 xenograft model, where the cells were inoculated into J:nu mice in a manner identical to the DAOY cell line. In the D341 model, most mice developed spinal metastasis that was detectable by IVIS imaging within 7 days of inoculation. We thus tested whether (BMN)NPs and free BMN-673 were efficacious against existing spinal metastasis by starting treatment at day 7 in the presence of leptomeningeal metastasis (Fig 8A). 4 repeat doses of (BMN)NPs (0.25 mg/kg) were efficacious in reducing cerebral and spinal tumor burden in the D341 model, whereas the free BMN-673 treatment (at an equitoxic dose to the NPs) showed no effect. We sought to improve this therapy with a temozolomide (TMZ) combination approach. The synergy between TMZ and PARPi is well documented in vitro, as PARP1 trapping sensitizes cells to the DNA methylation mechanism of TMZ (37, 38). However, in vivo, a reduction in the PARPi dose is necessary to prevent acute toxicity (39). The (BMN)NP and TMZ combination was well-tolerated in mice without a necessary dose-reduction and led to tumor clearance in 4 out of 6 mice tested (Fig 8B) with similar extents of mean weight-loss across all treatment groups (Fig 8C). We were unable to test the synergy between free BMN-673 and TMZ, as the combination was not tolerated at even the lowest doses of BMN-673 (0.03 mg/kg). Overall, these results imply that intra-CSF delivery of nanoparticle encapsulated drugs is a viable treatment regimen for MB with a meaningful advantage over intrathecal administration of free drug alone in terms of pronounced and sustained in vivo activity.
Figure 8 |. Tumor inhibition and anti-metastatic capacity of (BMN)NPs + TMZ in D341 tumor model mice:

(A) Whole body bioluminescence images of J:Nu mice inoculated with D341 cells over 4 weeks. Bioluminescence scale (p/sec/cm2/sr) is different for first 2 rows versus last 2 rows. (B) Survival curves for BMN-673 treated (n=7), (BMN)NP treated (n=9), (BMN)NP + TMZ treated (n=8), and control mice (n =8). Log-rank test, followed by repeated-measures ANOVA **** P <0.0001. (C) Change in body weights of all groups. Data presented as mean +/− s.d.
DISCUSSION
Direct infusion of drugs into the CSF has emerged as a promising method of circumventing the BBB in the treatment of MB (13). Given that leptomeningeal recurrence remains the leading cause of mortality in patients, there is a compelling rationale for an administration route that increases intra-CSF drug exposure, and several ongoing clinical trials are leveraging this pathway (40–43). Despite the potential advantages of intra-CSF delivery, maintaining high drug concentration in the subarachnoid space in the presence of rapid CSF flow and rapid clearance of CSF macromolecules remains a challenge. Here, we describe a PLA-HPG NP platform that persists in the subarachnoid region for long periods of time, in contrast to the fate of freely administered small molecules. Here, we demonstrate long-term retention of the NPs in the CSF space of tumor-free mice, as well as preferential accumulation and retention in CSF-adjacent tumors, using PET/CT with [89Zr]Zr-DFO-NP and Cy5-NP fluorescent whole-body imaging. We were encouraged by these findings, as increasing accumulation of NPs in the tumor early-on in CSF circulation lessens the probability of mononuclear phagocytic system and renal system clearance.
The vast majority of polymeric NPs exhibit substantial absorption in the spleen and drug clearance organs such as the liver and kidney, potentially limiting their therapeutic use (28, 44). Even with functionalized BBB penetrating modalities, NP accumulation in the brain when delivered systemically is typically limited to less than 1% of total activity, and bulk of the delivered NPs collect into the spleen and liver (45). Previous work suggested that when HPG-coated NPs are delivered systemically, they exhibit reduced recognition and clearance by the reticuloendothelial system compared with other commonly used polymeric NPs (24, 46). Hence, we predicted that PLA-HPG NPs would retain their ideal properties in the CSF space as well, with the hyperbranched surface structure forming a steric barrier around the NPs that extends circulation time and allows for greater tumor site accumulation.
To quantify the whole-body biodistribution of our NPs in healthy and tumor-bearing mice, we used PET imaging, which is a non-invasive tool with excellent quantitative capability, and high detection sensitivity. By leveraging the benefits of 89Zr-labeling (such as stability, long half-life, and well-characterized chelating properties), our present PET methods may readily extend to imaging studies in humans. [89Zr]Zr-DFO-NP showed less than 15% accumulation in clearance organs at all time points as measured by image-based quantification. We did observe a peak in the bladder as early as 5 m post injection, but bladder uptake is unlikely to be due to extravasated NPs. The size limitation of the kidney glomerular filtration is 48 kDa, or 5 nm for polymers such as PEG and dextran, and it is improbable that NPs degrade rapidly enough in the first 5 min after delivery to accumulate in the bladder. Furthermore, comparable peaks with [89Zr]Zr-DFO indicated it was not from extravasated NPs. The highest non-CNS accumulation occurred in the cervical lymph nodes, and not the liver or spleen or bone marrow. This observation is in line with previous reports on the drainage of CSF injected tracers (47, 48). Additionally, we observed greater transfer from CSF to systemic clearance in tumor bearing mice compared to healthy mice. This may be due to an abnormally leaky vasculature with the tumor microenvironment and/or dysfunctional lymphatic drainage toward brain draining lymph nodes and will be an area of further investigation.
We also observed accumulation of the radiolabeled NPs in the meninges of tumor-bearing mice, as well as in the tumor foci in the cerebellum. It is unclear whether the NPs are engulfed by tumor-associated immune cells in the brain before trafficking to the meninges, or whether NPs arrive at the meninges and are then taken up by resident immune-cell types. It is likely a combination of both pathways: 1) bulk CSF flow draining across the meninges and through the lymphatic system to the cervical lymph nodes, and 2) trafficking of NP-associated immune cells that represent potential NP carrier cells toward tumor-draining lymph nodes. In this respect, parenchymal border macrophages of the perivascular spaces and MHC-II+ meningeal macrophages have recently been demonstrated to play crucial roles in CSF flow dynamics and meningeal immune surveillance, respectively (49, 50). There is a portion of NPs in the tumor microenvironment that were not associated with either macrophages or microglia – these NPs may have been taken up by MB tumor cells specifically, or by other cells in the brain parenchyma such as astrocytes. The transport of NPs by the bulk flow of CSF also contributed to the lack of notable difference in CSF accumulation based on NP surface properties. This finding suggests that the flowing CSF environment reduces opportunities for interactions with cells and free-floating proteins.
Through PET imaging, we were able to detect presence of NPs in the CNS at 12 days, with a reduced but still detectable presence even at 21 days, which was the maximum timepoint we could assess due to limitations imposed by isotope half-life and PET imaging sensitivity. This long-term retention of NPs, combined with controlled drug release, offers the potential for prolonged drug exposure at the tumor site, and a means of improving the overall half-life of drug following intra-CSF administration. Given the well-studied biodegradation of PLA in biomedical systems (51), we predict that all of the NPs will disappear by ~6 months. In addition, because activity in the CNS represents at least 75% of total ID/g at all time points measured, there is a greatly reduced risk of widespread systemic toxicity. As proof-of-concept, we loaded our NPs with a promising new class of drugs, PARP inhibitors, that are limited by BBB penetration and cause widespread toxicity. PARPi were initially developed to sensitize tumor cells to conventional DNA damaging agents, and increasing evidence shows that PARPi are effective at sensitizing cells to radiotherapy, temozolomide, and topoisomerase poisons and inhibitors (52, 53). However, efforts in clinic to use PARPi in combination therapies have been marred by the high toxicity profile, and no PARPi has been approved for use in combination (14, 54). For pediatric CNS tumors, veliparib is the most clinically advanced PARPi due to its ability to cross the BBB. It has been evaluated in combination with temozolomide and with temozolomide plus radiotherapy, but difficulty in dose-escalation without causing toxicity and the absence of a survival benefit have slowed progress in the clinic (55, 56). Recent developments in our mechanistic understanding of PARPi sensitization have revealed that TMZ sensitization is induced by PARP1 trapping, which is consistent with the failure of veliparib, which has relatively poor PARP1 trapping ability (57, 58). In comparison, talazoparib (BMN-673) is a potent PARP1 trapper but is constrained by its inability to bypass the BBB in meaningful quantities.
Our study has several limitations. First, the evaluation of drug release kinetics was validated through in vitro assays, which may not fully represent the actual drug release dynamics after injection in vivo. To obtain a comprehensive understanding of the detectable drug concentrations in the CSF at different time points, further experiments in larger animal models with larger CSF volumes, which allow for sampling over time, will be necessary. In addition, the in vivo therapeutic efficacy of BMN-NPs was conducted using only two models of MB, which may not sufficiently capture the heterogeneity present within MB subtypes. To establish the clinical applicability of our therapeutic approach, future efficacy studies utilizing larger patient-derived xenograft (PDX) panels will be necessary. Furthermore, it should be noted that the reliance on mouse models in this study may not fully encompass the physiological characteristics of the CSF environment and leptomeningeal dissemination in medulloblastoma in humans.
Our studies with BMN-673 represent the first preclinical study of administering PARPi intrathecally, and we observed toxicity of the free drug both alone and in combination with TMZ. Our data suggest that delivery of NPs directly into the CSF likely prolongs the presence of NPs and hence drug exposure to cancer cells, resulting in favorable anti-tumor effects with minimal damage in healthy tissue. In the clinic, repeat administration of NPs, perhaps through an Ommaya reservoir, an intraventricular catheter used for delivery of drugs into the CSF, may improve therapeutic efficacy as compared with either systemic administration or free drug administration. We anticipate that this integrated treatment approach could open new investigations in PARPi or other drug combination therapies without compromising tolerability. In addition, this approach could lead to promising avenues of treatment for other diseases associated with extensive LMS, such as leptomeningeal metastases from primary malignancies such as lung, breast, and melanoma cancer. Future investigations will focus on exploring the efficacy of drug-loaded NPs in a leptomeningeal metastasis model, as well as on optimizing the synergistic combination of NPs with ionizing radiation and other DNA-damaging agents, such as topoisomerase poisons.
Materials and Methods
Study design
The aim of this study was to develop nanoparticles that encapsulate PARPi and to evaluate their efficacy in preclinical models of medulloblastoma. First, we characterized NPs and the effect of size, charge, surface chemistry on uptake and retention in the CNS, before evaluating biodistribution through PET imaging of healthy and tumor-bearing mice. Having determined optimal NP settings, we then determined dosage of free drug and NP to be used for preclinical studies. We then evaluated the impacts of single dosing, repeated dosing, and synergistic dosing of (BMN)NPs in our preclinical model (J:nu mice inoculated with luciferase-expressing DAOY cells) in vivo.
Replicates varied depending on experiment. Synthesized NP characterization was done in technical triplicates, and the effect of NP size, charge, and surface chemistry on uptake in the leptomeningeal and perivascular space after i.c.m. administration of dye-conjugated PLA-HPG NPs had at least biological triplicates (n=6 for 3 h, 24 h, 5d, 7 d). Evaluation through imaging of brain and spinal cord sections under fluorescence microscopy after 24 h and 48 h involved taking 10–15 representative slices per mouse (n=5). Evaluation of NPs in the CNS compartment in healthy mice by PET activity was done over 2 hours (n=2), while whole body biodistribution was observed over 12 days (n=4). These experiments were repeated in tumor-bearing mice (n=2 for CNS compartment evaluations, n=4 for whole body distribution). Cy5-NP quantification in the brain was evaluated 24 hours after i.c.m. administration between healthy and tumor-bearing mice (n=6 per group). Preliminary NP and free-drug dosage (n=1) was done before evaluating MTD for free drug and NPs at day 3 and 7 (n=6 per group). Survival with single dose was evaluated over 17 weeks (n=5 for control, n=5 for free drug, and n=6 for NPs). Changes in body weight were tracked, along with relative bioluminescence intensity of whole brain to evaluate tumor. Repeat dosing was similarly evaluated (n=9 for control, n=7 for free drug, n=8 for NP). Lastly, synergistic treatment of (BMN)NP and TMZ was evaluated (n=8 for control, n=7 for BMN free drug, n=9 for (BMN)NP, n=8 for (BMN)NP+TMZ). All animal procedures and protocols were approved by the Yale University Institutional Animal Care and Use Committee. Mice were grouped such that the average weight would be similar across the different conditions. Conditions were assigned randomly to groups. Study was not blinded.
Human medulloblastoma cell lines
DAOY, D341, and D283 were purchased from the American Type Culture Collection (ATCC) and cultured following supplier instructions.
Mouse lines
All procedures were approved by the Yale University Institutional Animal Care and Use Committee and performed in accordance with the guidelines and policies of the Yale Animal Resource Center. BALB/C mice (Charles River, 8 weeks, female) or J:Nu mice (Jackson Labs, 8 weeks, female) were used in all studies unless otherwise indicated.
Intracisterna magna (i.c.m.) injection and catheter implantation
Mice were anesthetized with ketamine/xylazine i.p., and ophthalmic solution placed on the eyes to prevent drying and the head of the mouse while secured in a stereotaxic frame. After making a skin incision, the muscle layers were retracted and the CM exposed. For bolus injection, a Hamilton syringe (coupled to a 33-gauge needle) was used to deliver the volume of the desired solution into the CSF-filled CM compartment. the needle was left in place for 1–2 m to avoid backflow. Mice were then glued with Vet bond, sutured and allowed to recover on a heating pad until active.
Catheter implantation was done with 32G IT catheter (0046EO; ReCathCo). For mice implanted with catheters, the trimmed end of the catheter was inserted into the cisterna magna, fixed along the superficial lateral muscles with tissue glue (Histoacryl), and the outer muscle layers were sutured. The mouse was allowed to recover on a heating pad until active, and local anesthesia ointment was used on the wound. Animals were also given buprenorphine (0.06 mg/kg every 12 h) for pain management. For 3 d after surgery, injection of buprenorphine was repeated, and mice were monitored for any signs of pain and/or distress. For any administration with the catheter, a 27G needle was fitted to the outer catheter tube and used to deliver up to 6 μL of NP or drug.
Statistical analysis
Data analysis and visualization was performed using Prism 7.0 (GraphPad Software). Graphs represent either group mean values ± s.d. (as indicated in the figure legends) or individual values. P values were calculated with log-rank statistics for survival analyses, then repeated-measures analysis of variance (ANOVA). A one-way ANOVA was used for multiple comparisons with Dunnett’s post hoc test for group-wise comparisons unless otherwise stated. Mann-Whitney U test was used for comparison between two groups. P < 0.05 was considered statistically significant. P values are denoted with asterisks: P > 0.05, ns; *P < 0.05; **P < 0.01; ***P < 0.001; and ****P < 0.0001. Individual- data for experiments where n < 20 are provided in data file S1.
Supplementary Material
Funding
This work was supported by grants from the National Institutes of Health (CA149128 to WMS and R01CA215453 to RSB, and R01NS13005701 to JLT and LG), the National Brain Tumor Society DDR-C grant (to RSB), and by the Yale Cancer Center Discovery Fund (to WMS and RSB).
Footnotes
Competing interests
MK, RSB, and WMS are inventors on a patent application describing intrathecal administration of nanoparticles for treating cancer (Title: INTRATHECAL NANOPARTICLE DELIVERY FOR TREATMENT OF LEPTOMENINGEAL TUMORS, U.S. Patent Application No. 63/383,211). RSB and WMS are co-founders of B3 Therapeutics. WMS is a consultant to Xanadu Bio, B3 Therapeutics, Stradefy Biosciences, Johnson & Johnson, Celanese, Cranius, and CMC Pharma. RSB is a consultant to Cybrexa therapeutics, Alphina therapeutics, Modifi bio, Sage Biosciences (SAB), and Aprea (SAB). The rest of the authors declare that they have no competing interests.
Data and materials availability
All data associated with this study are present in the paper or supplementary materials. Requests for data should be addressed to WMS. Reagents used to conduct these experiments are either publicly available as detailed in the materials section or available with appropriate materials transfer agreements. PLA-HPG polymers synthesized for this project are available from WMS under a materials transfer agreement with Yale University.
References
- 1.Rutkowski S, Cohen B, Finlay J, Luksch R, Ridola V, Valteau-Couanet D, Hara J, Garre ML, Grill J, Medulloblastoma in young children, Pediatr. Blood Cancer 54, 635–637 (2010). [DOI] [PubMed] [Google Scholar]
- 2.Gilbertson RJ, Ellison DW, The origins of medulloblastoma subtypes, Annu. Rev. Pathol. 3, 341–365 (2008). [DOI] [PubMed] [Google Scholar]
- 3.Kann BH, Park HSM, Lester-Coll NH, Yeboa DN, Benitez V, Bindra RS, Khan AJ, Marks AM, Roberts KB, Adjuvant Radiation Therapy Patterns and Survival Implications for Medulloblastoma in Young Children, Int. J. Radiat. Oncol 96, 1574–1581 (2016). [DOI] [PubMed] [Google Scholar]
- 4.Koschmann C, Bloom K, Upadhyaya S, Geyer JR, Leary SES, Survival After Relapse of Medulloblastoma, J. Pediatr. Hematol. Oncol. 38, 269–273 (2016). [DOI] [PubMed] [Google Scholar]
- 5.Zapotocky M, Mata-Mbemba D, Sumerauer D, Liby P, Lassaletta A, Zamecnik J, Krskova L, Kyncl M, Stary J, Laughlin S, Arnoldo A, Hawkins C, Tabori U, Taylor MD, Bouffet E, Raybaud C, Ramaswamy V, Differential patterns of metastatic dissemination across medulloblastoma subgroups, J. Neurosurg. Pediatr. 21, 145–152 (2018). [DOI] [PubMed] [Google Scholar]
- 6.Fujimoto T, Pharmacokinetics of intrathecal chemotherapy and clinical problems, Japanese J. Cancer Chemother. 11, 1536–1542 (1984). [PubMed] [Google Scholar]
- 7.Chatelut E, Kim T, Kim S, A slow-release methotrexate formulation for intrathecal chemotherapy, Cancer Chemother. Pharmacol. 32, 179–182 (1993). [DOI] [PubMed] [Google Scholar]
- 8.Esteva FJ, Soh LT, Holmes FA, Plunkett W, Meyers CA, Forman AD, Hortobagyi GN, Phase II trial and pharmacokinetic evaluation of cytosine arabinoside for leptomeningeal metastases from breast cancer, Cancer Chemother. Pharmacol. 46, 382–386 (2000). [DOI] [PubMed] [Google Scholar]
- 9.Zhou J, Patel TR, Sirianni RW, Strohbehn G, Zheng MQ, Duong N, Schafbauer T, Huttner AJ, Huang Y, Carson RE, Zhang Y, Sullivan DJ, Piepmeier JM, Saltzman WM, Highly penetrative, drug-loaded nanocarriers improve treatment of glioblastoma, Proc. Natl. Acad. Sci. U. S. A. 110, 11751–11756 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Shyam R, Ren Y, Lee J, Braunstein KE, Mao HQ, Wong PC, Intraventricular delivery of siRNA nanoparticles to the central nervous system, Mol. Ther. - Nucleic Acids 4, e242 (2015). [DOI] [PubMed] [Google Scholar]
- 11.Saucier-Sawyer JK, Seo YE, Gaudin A, Quijano E, Song E, Sawyer AJ, Deng Y, Huttner A, Saltzman WM, Distribution of polymer nanoparticles by convection-enhanced delivery to brain tumors, J. Control. Release 232, 103–112 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Gu X, Song Q, Zhang Q, Huang M, Zheng M, Chen J, Wei D, Chen J, Wei X, Chen H, Zheng G, Gao X, Clearance of two organic nanoparticles from the brain via the paravascular pathway, J. Control. Release 322, 31–41 (2020). [DOI] [PubMed] [Google Scholar]
- 13.Khang M, Bindra RS, Mark Saltzman W, Intrathecal delivery and its applications in leptomeningeal disease, Adv. Drug Deliv. Rev. 186, 114338 (2022). [DOI] [PubMed] [Google Scholar]
- 14.Curtin NJ, Szabo C, Poly(ADP-ribose) polymerase inhibition: past, present and future, Nat. Rev. Drug Discov. 19, 711–736 (2020). [DOI] [PubMed] [Google Scholar]
- 15.van Vuurden DG, Hulleman E, Meijer OLM, Wedekind LE, Kool M, Witt H, Peter Vandertop PW, Würdinger T, Noske DP, Kaspers GJL, Cloos J, PARP inhibition sensitizes childhood high grade glioma, medulloblastoma and ependymoma to radiation, Oncotarget 2, 984–986 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Buck J, Dyer PJC, Hii H, Carline B, Kuchibhotla M, Byrne J, Howlett M, Whitehouse J, Ebert MA, McDonald KL, Gottardo NG, Endersby R, Veliparib Is an Effective Radiosensitizing Agent in a Preclinical Model of Medulloblastoma, Front. Mol. Biosci 8, 633344 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Song E, Gaudin A, King AR, Seo YE, Suh HW, Deng Y, Cui J, Tietjen GT, Huttner A, Saltzman WM, Surface chemistry governs cellular tropism of nanoparticles in the brain, Nat. Commun. 8, 15322 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Deng Y, Yang F, Cocco E, Song E, Zhang J, Cui J, Mohideen M, Bellone S, Santin AD, Saltzman WM, Improved i.p. drug delivery with bioadhesive nanoparticles, Proc. Natl. Acad. Sci. U. S. A. 113, 11453–11458 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Wang Y, Malik S, Suh HW, Xiao Y, Deng Y, Fan R, Huttner A, Bindra RS, Singh V, Saltzman WM, Bahal R, Anti-seed PNAs targeting multiple oncomiRs for brain tumor therapy, Sci. Adv. 9, eabq7459 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Tong R, Coyle VJ, Tang L, Barger AM, Fan TM, Cheng J, Polylactide nanoparticles containing stably incorporated cyanine dyes for in vitro and in vivo imaging applications, Microsc. Res. Tech. 73, 901–909 (2010). [DOI] [PubMed] [Google Scholar]
- 21.Iliff JJ, Wang M, Liao Y, Plogg BA, Peng W, Gundersen GA, Benveniste H, Vates GE, Deane R, Goldman SA, Nagelhus EA, Nedergaard M, A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, LQFOXGLQJDP\ORLGȕ, Sci. Transl. Med. 4, 147ra111 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Iliff JJ, Lee H, Yu M, Feng T, Logan J, Nedergaard M, Benveniste H, Brain-wide pathway for waste clearance captured by contrast-enhanced MRI, J. Clin. Invest. 123, 1299–1309 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Armstrong JK, Wenby RB, Meiselman HJ, Fisher TC, The hydrodynamic radii of macromolecules and their effect on red blood cell aggregation, Biophys. J. 87, 4259–4270 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Deng Y, Saucier-Sawyer JK, Hoimes CJ, Zhang J, Seo YE, Andrejecsk JW, Saltzman WM, The effect of hyperbranched polyglycerol coatings on drug delivery using degradable polymer nanoparticles, Biomaterials 35, 6595–6602 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Deng Y, Ediriwickrema A, Yang F, Lewis J, Girardi M, Saltzman WM, A sunblock based on bioadhesive nanoparticles, Nat. Mater. 14, 1278–1285 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Hu JK, Suh HW, Qureshi M, Lewis JM, Yaqoob S, Moscato ZM, Griff S, Lee AK, Yin ES, Mark Saltzman W, Girardi M, Nonsurgical treatment of skin cancer with local delivery of bioadhesive nanoparticles, Proc. Natl. Acad. Sci. U. S. A. 118, e2020575118 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Mohideen M, Quijano E, Song E, Deng Y, Panse G, Zhang W, Clark MR, Saltzman WM, Degradable Bioadhesive Nanoparticles for Prolonged Intravaginal Delivery and Retention of Elvitegravir, Biomaterials 144, 144–154 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Wilhelm S, Tavares AJ, Dai Q, Ohta S, Audet J, Dvorak HF, Chan WCW, Analysis of nanoparticle delivery to tumours, Nat. Rev. Mater. 1, 16014 (2016). [Google Scholar]
- 29.Walkey CD, Olsen JB, Guo H, Emili A, Chan WCW, Nanoparticle size and surface chemistry determine serum protein adsorption and macrophage uptake, J. Am. Chem. Soc. 134, 2139–2147 (2012). [DOI] [PubMed] [Google Scholar]
- 30.De Jong WH, Hagens WI, Krystek P, Burger MC, Sips AJAM, Geertsma RE, Particle size-dependent organ distribution of gold nanoparticles after intravenous administration, Biomaterials 29, 1912–1919 (2008). [DOI] [PubMed] [Google Scholar]
- 31.Deri MA, Zeglis BM, Francesconi LC, Lewis JS, PET Imaging with 89Zr: From Radiochemistry to the Clinic, Nucl. Med. Biol 40, 3–14 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Bernards CM, Luger TJ, Malmberg AB, Hill HF, Yaksh TL, Liposome encapsulation prolongs alfentanil spinal analgesia and alters systemic redistribution in the rat, Anesthesiology 77, 529–535 (1992). [DOI] [PubMed] [Google Scholar]
- 33.Angst MS, Drover DR, Pharmacology of drugs formulated with DepoFoam: a sustained release drug delivery system for parenteral administration using multivesicular liposome technology, Clin. Pharmacokinet 45, 1153–1176 (2006). [DOI] [PubMed] [Google Scholar]
- 34.Musumeci T, Ventura CA, Giannone I, Ruozi B, Montenegro L, Pignatello R, Puglisi G, PLA/PLGA nanoparticles for sustained release of docetaxel, Int. J. Pharm. 325, 172–179 (2006). [DOI] [PubMed] [Google Scholar]
- 35.Danafar H, Rostamizadeh K, Davaran S, Hamidi M, PLA-PEG-PLA copolymer-based polymersomes as nanocarriers for delivery of hydrophilic and hydrophobic drugs: preparation and evaluation with atorvastatin and lisinopril, Drug Dev. Ind. Pharm. 40, 1411–1420 (2014). [DOI] [PubMed] [Google Scholar]
- 36.Dong Y, Feng SS, Methoxy poly(ethylene glycol)-poly(lactide) (MPEG-PLA) nanoparticles for controlled delivery of anticancer drugs, Biomaterials 25, 2843–2849 (2004). [DOI] [PubMed] [Google Scholar]
- 37.Gupta SK, Smith EJ, Mladek AC, Tian S, Decker PA, Kizilbash SH, Kitange GJ, Sarkaria JN, PARP inhibitors for sensitization of alkylation chemotherapy in glioblastoma: Impact of blood-brain barrier and molecular heterogeneity, Front. Oncol 9, 670 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Hopkins TA, Ainsworth WB, Ellis PA, Donawho CK, DiGiammarino EL, Panchal SC, Abraham VC, Algire MA, Shi Y, Olson AM, Johnson EF, Wilsbacher JL, Maag D, PARP1 trapping by PARP inhibitors drives cytotoxicity in both cancer cells and healthy bone marrow, Mol. Cancer Res. 17, 409–419 (2019). [DOI] [PubMed] [Google Scholar]
- 39.Baldwin P, Likhotvorik R, Baig N, Cropper J, Carlson R, Kurmasheva R, Sridhar S, Nanoformulation of Talazoparib Increases Maximum Tolerated Doses in Combination With Temozolomide for Treatment of Ewing Sarcoma, Front. Oncol 9, 1416 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Pandit-Taskar N, Zanzonico PB, Kramer K, Grkovski M, Fung EK, Shi W, Zhang Z, Lyashchenko SK, Fung AM, Pentlow KS, Carrasquillo JA, Lewis JS, Larson SM, Cheung NKV, Humm JL, Biodistribution and dosimetry of intraventricularly administered 124I-omburtamab in patients with metastatic leptomeningeal tumors, J. Nucl. Med. 60, 1794–1801 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Kramer K, Pandit-Taskar N, Humm JL, Zanzonico PB, Haque S, Dunkel IJ, Wolden SL, Donzelli M, Goldman DA, Lewis JS, Lyashchenko SK, Khakoo Y, Carrasquillo JA, Souweidane MM, Greenfield JP, Lyden D, De Braganca KD, Gilheeney SW, Larson SM, Cheung NKV, A phase II study of radioimmunotherapy with intraventricular 131I-3F8 for medulloblastoma, Pediatr. Blood Cancer 65, e26754 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Okada K, Soejima T, Sakamoto H, Hirato J, Hara J, Phase II study of reduced-dose craniospinal irradiation and combination chemotherapy for children with newly diagnosed medulloblastoma: A report from the Japanese Pediatric Brain Tumor Consortium, Pediatr. Blood Cancer 67, e28572 (2020). [DOI] [PubMed] [Google Scholar]
- 43.Naung H, Cohen KJ, An intrathecal limited postoperative chemotherapy regimen for the treatment of young children with nodular/desmoplastic medulloblastoma and medulloblastoma with extensive nodularity, J. Neurooncol. 152, 567–572 (2021). [DOI] [PubMed] [Google Scholar]
- 44.Cheng CJ, Tietjen GT, Saucier-Sawyer JK, Saltzman WM, A holistic approach to targeting disease with polymeric nanoparticles, Nat. Rev. Drug Discov. 14, 239–247 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Zhou Y, Peng Z, Seven ES, Leblanc RM, Crossing the blood-brain barrier with nanoparticles, J. Control. Release 270, 290–303 (2018). [DOI] [PubMed] [Google Scholar]
- 46.Saucier-Sawyer JK, Deng Y, Seo YE, Cheng CJ, Zhang J, Quijano E, Saltzman WM, Systemic Delivery of Blood-Brain Barrier Targeted Polymeric Nanoparticles Enhances Delivery to Brain Tissue, J. Drug Target 23, 736–749 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Louveau A, Smirnov I, Keyes TJ, Eccles JD, Rouhani SJ, Peske JD, Derecki NC, Castle D, Mandell JW, Lee KS, Harris TH, Kipnis J, Structural and functional features of central nervous system lymphatic vessels, Nature 523, 337–341 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Aspelund A, Antila S, Proulx ST, Karlsen TV, Karaman S, Detmar M, Wiig H, Alitalo K, A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules, J. Exp. Med. 212, 991–999 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Drieu A, Du S, Storck SE, Rustenhoven J, Papadopoulos Z, Dykstra T, Zhong F, Kim K, Blackburn S, Mamuladze T, Harari O, Karch CM, Bateman RJ, Perrin R, Farlow M, Chhatwal J, Brosch J, Buck J, Farlow M, Ghetti B, Adams S, Barthelemy N, Benzinger T, Brandon S, Buckles V, Cash L, Chen C, Chua J, Cruchaga C, Denner D, Dincer A, Donahue T, Fagan A, Feldman B, Flores S, Franklin E, Joseph-Mathurin N, Gonzalez A, Gordon B, Gray J, Gremminger E, Groves A, Hassenstab J, Hellm C, Herries E, Hoechst-Swisher L, Holtzman D, Hornbeck R, Jerome G, Keefe S, Koudelis D, Li Y, Marsh J, Martinez R, Mawuenyega K, McCullough A, McDade E, Morris J, Norton J, Shady K, Sigurdson W, Smith J, Wang P, Wang Q, Xiong C, Xu J, Xu X, Allegri R, Mendez PC, Egido N, Araki A, Ikeuchi T, Ishii K, Kasuga K, Bechara J, Brooks W, Schofield P, Berman S, Goldberg S, Ikonomovic S, Klunk W, Lopez O, Mountz J, Nadkarni N, Patira R, Smith L, Snitz B, Thompson S, Weamer E, Bodge C, Salloway S, Carter K, Duong D, Johnson E, Levey A, Ping L, Seyfried NT, Fitzpatrick C, Chui H, Ringman J, Day GS, Graff-Radford N, Graham M, Stephens S, La Cruz CD, Goldman J, Mejia A, Neimeyer K, Noble J, Diffenbacher A, Yakushev I, Levin J, Vöglein J, Douglas J, Fox N, Grilo M, Mummery C, O’Connor A, Esposito B, Goate A, Renton A, Fujii H, Senda M, Shimada H, Gardener S, Martins R, Sohrabi H, Taddei K, Gräber-Sultan S, Häsler L, Hofmann A, Jucker M, Käser S, Kuder-Buletta E, Laske C, Preische O, Haass C, Morenas-Rodriguez E, Nuscher B, Ihara R, Nagamatsu A, Niimi Y, Jack C, Koeppe R, Mason NS, Masters C, Obermüller U, Hu S, Randolph GJ, Smirnov I, Kipnis J, Parenchymal border macrophages regulate the flow dynamics of the cerebrospinal fluid, Nature 611, 585–593 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Rebejac J, Eme-Scolan E, Arnaud Paroutaud L, Kharbouche S, Teleman M, Spinelli L, Gallo E, Roussel-Queval A, Zarubica A, Sansoni A, Bardin Q, Hoest P, Michallet MC, Brousse C, Crozat K, Manglani M, Liu Z, Ginhoux F, McGavern DB, Dalod M, Malissen B, Lawrence T, Rua R, Meningeal macrophages protect against viral neuroinfection, Immunity 55, 2103–2117.e10 (2022). [DOI] [PubMed] [Google Scholar]
- 51.Saltzman WM, Drug Delivery: Engineering Principles for Drug Therapy (Oxford University Press, 2001). [Google Scholar]
- 52.Lord CJ, Ashworth A, PARP inhibitors: Synthetic lethality in the clinic, Science. 355, 1152–1158 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Lu Y, Liu Y, Pang Y, Pacak K, Yang C, Double-barreled gun: Combination of PARP inhibitor with conventional chemotherapy, Pharmacol. Ther. 188, 168–175 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Lesueur P, Chevalier F, Austry JB, Waissi W, Burckel H, Noël G, Habrand JL, Saintigny Y, Joly F, Poly-(ADP-ribose)-polymerase inhibitors as radiosensitizers: A systematic review of pre-clinical and clinical human studies, Oncotarget 8, 69105–69124 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Su JM, Thompson P, Adesina A, Li XN, Kilburn L, Onar-Thomas A, Kocak M, Chyla B, McKeegan E, Warren KE, Goldman S, Pollack IF, Fouladi M, Chen A, Giranda V, Boyett J, Kun L, Blaney SM, A phase I trial of veliparib (ABT-888) and temozolomide in children with recurrent CNS tumors: A pediatric brain tumor consortium report, Neuro. Oncol. 16, 1661–1668 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Sim HW, Mcdonald KL, Lwin Z, Barnes EH, Rosenthal M, Foote MC, Koh ES, Back M, Wheeler H, Sulman EP, Buckland ME, Fisher L, Leonard R, Hall M, Ashley DM, Yip S, Simes J, Khasraw M, A randomized phase II trial of veliparib, radiotherapy, and temozolomide in patients with unmethylated MGMT glioblastoma: The VERTU study, Neuro. Oncol. 23, 1736–1749 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Murai J, Zhang Y, Morris J, Ji J, Takeda S, Doroshow JH, Pommier Y, Rationale for poly(ADP-ribose) polymerase (PARP) inhibitors in combination therapy with camptothecins or temozolomide based on PARP trapping versus catalytic inhibition, J. Pharmacol. Exp. Ther. 349, 408–416 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Murai J, Huang SYN, Das BB, Renaud A, Zhang Y, Doroshow JH, Ji J, Takeda S, Pommier Y, Trapping of PARP1 and PARP2 by clinical PARP inhibitors, Cancer Res. 72, 5588–5599 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Abou DS, Ku T, Smith-Jones PM, In vivo biodistribution and accumulation of 89Zr in mice, Nucl. Med. Biol 38, 675–681 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
All data associated with this study are present in the paper or supplementary materials. Requests for data should be addressed to WMS. Reagents used to conduct these experiments are either publicly available as detailed in the materials section or available with appropriate materials transfer agreements. PLA-HPG polymers synthesized for this project are available from WMS under a materials transfer agreement with Yale University.
