Convection-Enhanced Intraparenchymal Delivery of Drugs to the Brain

Number: 0731

Table Of Contents

Policy
Applicable CPT / HCPCS / ICD-10 Codes
Background
References


Policy

Scope of Policy

This Clinical Policy Bulletin addresses convection-enhanced intraparenchymal delivery of drugs to the brain.

Experimental and Investigational

Aetna considers convection-enhanced delivery of drugs into brain parenchyma experimental and investigational because the safety and effectiveness of this approach has not been established.


Table:

CPT Codes / HCPCS Codes / ICD-10 Codes

Code Code Description

Information in the [brackets] below has been added for clarification purposes.   Codes requiring a 7th character are represented by "+":

ICD-10 codes not covered for indications listed in the CPB (not all-inclusive):

C71.0 - C71.9 Malignant neoplasm of brain
C79.31, C79.49 Secondary malignant neoplasm of brain and other parts of nervous system
G30.0 - G30.9 Alzheimer's disease
G40.001 - G40.909 Epilepsy and recurrent seizures
R56.1 Post traumatic seizures
R56.9 Unspecified convulsions

Background

Convection-enhanced delivery (CED) is a method of directly administering drugs into the brain in order to enhance the distribution of drugs throughout the brain parehcyma.  It involves the stereotactic placement through cranial burr holes of several catheters into brain parenchyma and the subsequent infusion of antineoplastic agents or other therapeutic agents via a microinfusion pump.

This technique has been described in published pre-clinical and early clinical studies.  Although CED has been primarily evaluated for administering anti-neoplastics, this technique can be used for administering a wide range of agents, such as for treatment of Alzheimer’s disease and epilepsy.

Standard methods of local delivery of most drugs into the brain, either by intravenous injection and passage through the blood brain barrier (BBB), or intra-ventricular injection, has relied on diffusion, which results in a non-homogenous distribution of most agents.  Intravenous administration of drugs to the brain has been hampered by the BBB, which prevents the passage of large molecules.  The BBB is characterized by tight junctions between vascular endothelial cells, which prevent or impede various naturally occurring and synthetic substances (including anti-cancer drugs) from entering the brain.

Blood brain barrier disruption (BBBD) techniques have been used in chemotherapy of brain tumors to disrupt the BBB, in order to increase the concentration of chemotherapy drugs delivered to the tumor and prolong the drug-tumor contact time.  Chemotherapy may be administered in conjunction with mannitol to cause osmotic disruption of the BBB, or disruption may be attempted through other mechanisms with other substances.

Although BBBD techniques have been used for over 20 years, the efficacy of this technique has been questioned (ICSI, 2001; CMS, 2007), as the long-term effects of BBBD chemotherapy are unknown, and no randomized controlled trials have established the superiority of BBBD chemotherapy over conventional chemotherapy.  In addition, BBBD with chemotherapy is associated with a higher risk of complications than conventional chemotherapy.  Complications associated with BBBD include seizures, obtundation, focal neurologic deficits, cerebral herniation, strokes, death, as well as the side-effects related to the chemotherapeutic agents themselves.  Based upon a review of the evidence, CMS has issued a National Coverage Determination that stated that the use of osmotic BBBD is not reasonable and necessary when it is used as part of a treatment regimen for brain tumors (CMS, 2007).

In contrast to techniques that rely on diffusion, CED uses a pressure gradient established at the tip of an infusion catheter to push a drug into the extra-cellular space.  The intention is to distribute the drug more evenly, at higher concentrations, and over a larger area than when administered by diffusion alone.

Convection-enhanced delivery of anti-neoplastic agents may occur after craniotomy with tumor resection, once the patient is stable.  A separate hospital admission (apart from admission for tumor resection) is expected for CED for catheter placement and infusion of the therapeutic agent by means of a microinfusion pump.  Once the infusion is complete, the catheters are removed and the patient is discharged.

Novel, targeted anti-neoplastics are in development for brain tumors.  However, their administration has been hampered by the BBB, which prevents passage of large molecules.

One such targeted anti-neoplastic that is administered by CED is cintredekin besudotox, a novel cytotoxin-based therapy that is being investigated for the treatment of recurrent glioblastoma multiforme (GBM).  Cintredekin besudotox is a recombinant protein consisting of a single molecule composed of 2 parts:
  1. interleukin-13, which binds to receptors on tumor cells; and
  2. pseudomonas exotoxin (PE), a cytotoxin, which causes destruction of the tumor cell once the molecule is absorbed. 

Interleukin-13 receptors are present in substantial numbers on malignant glioma cells, but only a minimal amount on healthy brain cells.  Hence, cintredekin besudotox has the potential to target tumor cells, with minimal impact on surrounding normal brain tissue.

Because of its large size, cintredekin besudotox can not cross the BBB.  In clinical studies, cintredekin besudotox has been administered by CED.  Catheters are placed following tumor resection, in areas of microscopic tumor spread or at risk of tumor spread around the tumor resection cavity.  Because of the need to achieve homogenous distribution of cintredekin besudotox throughout the tumor infiltrated tissue, the catheters can not be placed in any previous resection cavity.

Once the patient is stable, approximately 2 weeks following craniotomy with tumor resection, the patient is admitted for catheter placement and anti-neoplastic infusion.  Catheters are strategically placed by neurosurgeons, taking into account the location of residual non-resectable tumor, brain anatomy, and fluid dynamics.  Anywhere from 2 to 4 catheters are placed during a surgical procedure lasting several hours.  Cintredekin besudotox is then slowly infused through the catheter directly into the brain over 96 hours.

Available phase I/II clinical studies of cintredekin besudotox suggest that this is a promising agent for treatment of recurrent glioblastoma multiforme.  A phase III trial (PRECISE) is currently underway.  Cintredekin besudotox has been granted fast-track development designation and orphan drug designation by the U.S. Food and Drug Administration (FDA), and may be approved by the FDA as early as 2007.

At present, the pharmacokinetics of CED are poorly understood (Sampson et al, 2006).  More research is needed to determine the optimal catheter location to distribute a drug to target tumor cells within the tumor mass and in the infiltrated adjacent parenchyma.  Optimal catheter design is being researched to minimize backflow, to maximize distribution in the brain, and to account for the need to maintain patient mobility.

Raghavan et al (2006) explained that, although CED has been under investigation since the early 1990’s, "this technique remains experimental because of both the absence of approved drugs for intraparenchymal delivery and the difficulty of guaranteed delivery to delineated regions of the brain."

Sampson et al (2008) determined the maximum tolerated dose (MTD), dose-limiting toxicity (DLT), and intra-cerebral distribution of a recombinant toxin (TP-38) targeting the epidermal growth factor receptor in patients with recurrent malignant brain tumors using the intra-cerebral infusion technique of CED.  A total of 20 patients were enrolled and stratified for dose escalation by the presence of residual tumor from 25 to 100 ng/ml in a 40-ml infusion volume.  In the last 8 patients, co-infusion of (123)I-albumin was performed to monitor distribution within the brain.  The MTD was not reached in this study.  Dose escalation was stopped at 100 ng/ml due to inconsistent drug delivery as evidenced by imaging the co-infused (123)I-albumin.  Two DLTs were seen, and both were neurological.  Median survival after TP-38 was 28 weeks (95 % confidence interval: 26.5 to 102.8).  Of the 15 patients treated with residual disease, 2 (13.3 %) demonstrated radiographical responses, including 1 patient with glioblastoma multiforme who had a nearly complete response and remains alive for over 260 weeks after therapy.  Co-infusion of (123)I-albumin demonstrated that high concentrations of the infusate could be delivered over 4 cm from the catheter tip.  However, only 3 of 16 (19 %) catheters produced intra-parenchymal infusate distribution, while the majority leaked infusate into the cerebrospinal fluid spaces.  Intra-cerebral CED of TP-38 was well-tolerated and produced some durable radiographical responses at doses less than or equal to 100 ng/ml.  The authors concluded that CED has significant potential for enhancing delivery of therapeutic macromolecules throughout the human brain.  However, the potential efficacy of drugs delivered by this technique may be severely constrained by ineffective infusion in many patients.

Fiandaca and colleagues (2008) stated that CED of substances within the human brain is becoming a more frequent experimental treatment option in the management of brain tumors, and more recently in phase 1 trials for gene therapy in Parkinson's disease (PD).  Benefits of this intracranial drug-transfer technology include a more efficient delivery of large volumes of therapeutic agent to the target region when compared with more standard delivery approaches (i.e., biopolymers, local infusion).  These researchers developed a reflux-resistant infusion cannula that allows increased infusion rates to be used.  They also described their efforts to visualize the CED process in vivo, using liposomal nanotechnology and real-time intra-operative MRI.  In addition to carrying the MRI contrast agent, nanoliposomes also provide a standardized delivery vehicle for the convection of drugs to a specific brain-tissue volume.  This technology provides an added level of assurance via visual confirmation of CED, allowing intra-operative alterations to the infusion if there is reflux or aberrant delivery.  These investigators proposed that these specific modifications to the CED technology will improve efficacy by documenting and standardizing the treatment-volume delivery.  Furthermore, they believe that this image-guided CED platform can be used in other translational neuroscience efforts, with eventual clinical application beyond neuro-oncology and PD.

In a review on novel drug delivery strategies in neuro-oncology, Bidros and Vogelbaum (2009) stated that an important impediment to finding effective treatments for malignant gliomas is the presence of the BBB, which serves to prevent delivery of potentially active therapeutic compounds.  Multiple efforts are focused on developing strategies to effectively deliver active drugs to brain tumor cells.  Convection-enhanced delivery and BBBD have emerged as leading investigational delivery techniques for the treatment of malignant brain tumors.  Clinical trials using these methods have been completed, with mixed results, and several more are being initiated.

Bidros et al (2010) stated that CED has emerged as a leading investigational delivery technique for the treatment of brain tumors.  Clinical trials utilizing these methods have been completed, with mixed results, and several more are being initiated.  However, the potential effectiveness of drugs delivered by CED may be severely constrained by poor durg distribution.

Sampson et al (2010) retrospectively analyzed the expected drug distribution based on catheter positioning data available from the CED arm of the PRECISE trial.  BrainLAB iPlan Flow software was used to estimate the expected drug distribution.  Only 49.8 % of catheters met all positioning criteria.  Still, catheter positioning score (hazard ratio 0.93, p = 0.043) and the number of optimally positioned catheters (hazard ratio 0.72, p = 0.038) had a significant effect on progression-free survival.  Estimated coverage of relevant target volumes was low, however, with only 20.1 % of the 2-cm penumbra surrounding the resection cavity covered on average.  Although tumor location and resection cavity volume had no effect on coverage volume, estimations of drug delivery to relevant target volumes did correlate well with catheter score (p < 0.003), and optimally positioned catheters had larger coverage volumes (p < 0.002).  Only overall survival (p = 0.006) was higher for investigators considered experienced after adjusting for patient age and Karnofsky Performance Scale score.  The authors concluded that potential effectiveness of drugs delivered by CED may be limited by ineffective delivery in many patients.

Buonerba et al (2011) stated that GBM is the most frequent and aggressive malignant glioma (MG), with a median survival time of 12 to15 months, despite current best treatment based on surgery, radiotherapy and systemic chemotherapy.  Many potentially active therapeutic agents are not effective by systemic administration, because they are unable to cross the BBB.  As intra-cerebral administration bypasses the BBB, it increases the number of drugs that can be successfully delivered to the brain, with the possibility of minor systemic toxicity and better effectiveness.  These researchers summarized the results of the extensive clinical research conducted on intra-cerebral therapy.  Biodegradable drug carriers, implantable subcutaneous reservoirs and CED represent the main techniques for intra-cerebral delivery, while conventional chemotherapy agents, radiolabeled antibodies and receptor-targeted toxins are the main classes of drugs for intra-cerebral therapy.  At the present time, biodegradable carmustine wafers, commercialized as Gliadel, are the only FDA-approved treatment for intra-cerebral chemotherapy of MG, but intra-cavitary delivery of mitoxantrone and radiolabeled anti-tenascin antibodies via implantable reservoirs has yielded promising results in uncontrolled trials.  The pressure-driven flow generated by CED can potentially distribute convected drugs over large volumes of the brain, independently on their intrinsic diffusivity.  Nevertheless, prominent technical problems, like back-flow, are yet to be properly addressed and contributed to the disappointing results of 2 phase III trials that investigated CED of cintredekin besudotox and TransMid in patients with recurrent GBM.

In a prospective, dose-escalation phase Ib study, Bruce et al (2011) examined the safety profile of topotecan via CED in the treatment of recurrent MGs and assessed radiographical response and survival.  Significant anti-tumor activity as described by radiographical changes and prolonged overall survival with minimal drug-associated toxicity was demonstrated.  A MTD was established for future phase II studies.  The authors concluded that topotecan by CED has significant anti-tumor activity at concentrations that are non-toxic to normal brain.  The potential for use of this therapy as a generally effective treatment option for MGs will be tested in subsequent phase II and phase III trials.

Lam et al (2011) stated that CED is a promising neurosurgical technique for the delivery of potential therapeutic agents to the PD-affected striatum.  Convection-enhanced delivery utilizes stereotactic insertion of a catheter to the striatum and continuous infusion to distribute agents in the brain parenchyma.  Insufficient attention to the details of CED may have contributed to early failures of translating candidate therapeutic agents from the laboratory to PD patients.  A literature review was performed to examine the factors that govern CED in the laboratory as well as translation in PD and these researchers found that although there have been significant developments in implant design, infusion parameters and infusate composition, there have not been enough comparative trials of different technologies.  Further optimization of CED is needed before it can be applied in the clinical setting and this will require a step-by-step breakdown of the different elements of delivery for independent testing.  The authors concluded that CED is a promising technique for delivering therapeutic agents to the striatum for the treatment of PD; but further refinements are necessary for successful clinical translation. 

Barua et al (2012) examined if  the peri-vascular distribution of solutes delivered by CED into the striatum of rats is affected by the molecular weight of the infused agent, by co-infusion of vasodilator, alteration of infusion rates or use of a ramping regime.  These investigators also wanted to make a preliminary comparison of the distribution of solutes with that of nanoparticles.  These researchers analysed the peri-vascular distribution of 4, 10, 20, 70, 150 kDa fluorescein-labelled dextran and fluorescent nanoparticles at 10 mins and 3 hrs following CED into rat striatum.  They investigated the effect of local vasodilatation, slow infusion rates and ramping on the peri-vascular distribution of solutes.  Co-localization with peri-vascular basement membranes and vascular endothelial cells was identified by immunohistochemistry.  The uptake of infusates by peri-vascular macrophages was quantified using stereological methods.  Widespread peri-vascular distribution and macrophage uptake of fluorescein-labelled dextran was visible 10 mins after cessation of CED irrespective of molecular weight.  However, a significantly higher proportion of peri-vascular macrophages had taken up 4, 10 and 20 kDa fluorescein-labelled dextran than 150 kDa dextran (p < 0.05, ANOVA).  Co-infusion with vasodilator, slow infusion rates and use of a ramping regime did not alter the peri-vascular distribution.  Convection-enhanced delivery of fluorescent nanoparticles indicated that particles co-localize with peri-vascular basement membranes throughout the striatum but, unlike soluble dextrans, are not taken up by peri-vascular macrophages after 3 hrs.  The authors concluded that the findings of this study suggested that widespread peri-vascular distribution and interaction with peri-vascular macrophages is likely to be an inevitable consequence of CED of solutes.  The potential consequences of peri-vascular distribution of therapeutic agents, and in particular cytotoxic chemotherapies, delivered by CED must be carefully considered to ensure safe and effective translation to clinical trials.

White et al (2012) described a single-center, phase I, dose-escalation clinical trial of carboplatin administered by CED to patients with recurrent or progressive GBM despite full standard treatment.  This trial will incorporate 6 cohorts of 3 patients each.  Cohorts will be treated in a sequential manner with increasing doses of carboplatin, subject to dose-limiting toxicity not being observed.  This protocol should facilitate the identification of the maximum-tolerated infused concentration of carboplatin by CED into the supratentorial brain.  This should facilitate the safe application of this technique in a phase II trial, treating patients with GBM, as well as for the treatment of other forms of malignant brain tumors, including metastases.

Anderson et al (2013) noted that CED for the treatment of malignant gliomas is a technique that can deliver chemotherapeutic agents directly into the tumor and the surrounding interstitium through sustained, low-grade positive-pressure infusion.  This allows for high local concentrations of drug within the tumor while minimizing systemic levels that often lead to dose-limiting toxicity.  Diffuse intrinsic pontine gliomas (DIPGs) are universally fatal childhood tumors for which there is currently no effective treatment.  In this report the authors described CED of the topoisomerase inhibitor topotecan for the treatment of DIPG in 2 children.  As part of a pilot feasibility study, the authors treated 2 pediatric patients with DIPG.  Stereotactic biopsy with frozen section confirmation of glial tumor was followed by placement of bilateral catheters for CED of topotecan during the same procedure.  The first patient underwent CED 210 days after initial diagnosis, after radiation therapy and at the time of tumor recurrence, with a total dose of 0.403 mg in 6.04 ml over 100 hours.  Her Karnofsky Performance Status (KPS) score was 60 before CED and 50 post-treatment.  Serial MRI initially demonstrated a modest reduction in tumor size and edema, but the tumor progressed and the patient died 49 days after treatment.  The second patient was treated 24 days after the initial diagnosis prior to radiation with a total dose of 0.284 mg in 5.30 ml over 100 hours.  Her KPS score was 70 before CED and 50 post-treatment.  Serial MRI similarly demonstrated an initial modest reduction in tumor size.  The patient subsequently underwent fractionated radiation therapy, but the tumor progressed and she died 120 days after treatment.  Topotecan delivered by prolonged CED into the brainstem in children with DIPG is technically feasible.  In both patients, high infusion rates (greater than 0.12 ml/hr) and high infusion volumes (greater than 2.8 ml) resulted in new neurological deficits and reduction in the KPS score, but lower infusion rates (less than 0.04 ml/hr) were well-tolerated.  While serial MRI showed moderate treatment effect, CED did not prolong survival in these 2 patients.  The authors concluded that more studies are needed to improve patient selection and determine the optimal flow rates for CED of chemotherapeutic agents into DIPG to maximize safety and efficacy.

Barua et al (2014) stated that CED describes a direct method of drug delivery to the brain through intra-parenchymal micro-catheters.  By establishing a pressure gradient at the tip of the infusion catheter in order to exploit bulk flow through the interstitial spaces of the brain, CED offers a number of advantages over conventional drug delivery methods -- bypass of the BBB, targeted distribution through large brain volumes and minimization of systemic side-effects.  Despite showing early promise, CED is yet to fulfill its potential as a mainstream strategy for the treatment of neurological diseases (e.g., Alzheimer's disease, high-grade glioma, and Parkinson's disease).  Substantial research effort has been dedicated to optimizing the technology for CED and identifying the parameters that govern successful drug distribution.  It seems likely that successful clinical translation of CED will depend on suitable catheter technology being used in combination with drugs with optimal physicochemical characteristics, and on neuropathological analysis in appropriate pre-clinical models.

Xia and colleagues (2014) examined a novel drug delivery system for the treatment of malignant brain gliomas; DOX complexed with nanodiamonds (ND-Dox), and administered via CED.  Drug retention and toxicity were examined in glioma cell lines, and distribution, retention and toxicity were examined in normal rat parenchyma.  Efficacy was assessed in a bioluminescence rodent tumor model.  NDs markedly enhanced DOX uptake and retention in glioma cells.  ND-Dox delivered via CED extended DOX retention and localized DOX toxicity in normal rodent parenchyma, and was significantly more efficient at killing tumor cells than uncomplexed DOX.  The authors concluded that outcomes from this work suggested that CED of ND-Dox is a promising approach for brain tumor treatment.

Aparicio-Blanco and Torres-Suarez (2015) stated that epidemiological data on central nervous system disorders call for a focus on the major hindrance to brain drug delivery, blood-central nervous system barriers. Otherwise, there is little chance of improving the short-term survival of patients with diseases such as GBM, which is one of the brain disorders associated with many years of life lost.  Targetable nano-carriers for treating malignant gliomas are a unique way to overcome low chemotherapeutic levels at target sites devoid of systemic toxicity.  These researchers described the currently available targetable nano-carriers, focusing particularly on one of the newest nano-carriers, lipid nano-capsules.  All of the strategies that are likely to be exploited by lipid nano-capsules to bypass blood-central nervous system barriers, including the most recent targeting approaches (mesenchymal cells), and novel administration routes (CED) were discussed, together with their most remarkable achievements in glioma-implanted animal models.  The authors concluded that although these systems are promising, much research remains to be done in this field.

Zhou and colleagues (2017) noted that CED is a technique designed to deliver drugs directly into the brain or tumors. Its ability to bypass the BBB has made it a promising drug delivery method for the treatment of primary brain tumors.  A number of clinical trials utilizing CED of various therapeutic agents have been conducted to treat patients with supra-tentorial high-grade gliomas.  Significant responses have been observed in certain patients in all of these trials.  However, the insufficient ability to monitor drug distribution and pharmacokinetics hampers CED from achieving its potentials on a larger scale.  Brainstem CED for DIPG treatment is appealing because this tumor is compact and has no definitive treatment.  The safety of brainstem CED has been established in small and large animals, and recently in early stage clinical trials.  There are a few current clinical trials of brainstem CED in treating DIPG patients using targeted macromolecules such as antibodies and immunotoxins.  Future advances for CED in DIPG treatment will come from several directions including: choosing the right agents for infusion; developing better agents and regimen for DIPG infusion; improving instruments and technique for easier and accurate surgical targeting and for allowing multi-session or prolonged infusion to implement optimal time sequence; and better understanding and control of drug distribution, clearance and time sequence.  The authors concluded that CED-based therapies for DIPG will continue to evolve with new understanding of the technique and the disease.

Jahangiri and colleagues (2017) noted that glioblastoma is the most common malignant brain tumor, and it carries an extremely poor prognosis.  Attempts to develop targeted therapies have been hindered because the BBB prevents many drugs from reaching tumors cells.  Furthermore, systemic toxicity of drugs often limits their therapeutic potential.  A number of alternative methods of delivery have been developed, one of which is CED.  The authors described CED as a therapeutic measure and review pre-clinical studies and the most prominent clinical trials of CED in the treatment of glioblastoma.  Moreover, they outlined numerous technical challenges that need to be met to overcome the issues encountered with the use of CED to treat glioblastoma to date.  Another consideration that will be important to prioritize going forward is that durable CED efficacy might require long-term convection at set intervals for months, as is often required for systemically administered chemotherapy to be effective for non-brain tumors.  The success of such a strategy may require implantable ports that can be cannulated to receive CED in an out-patient setting.  In addition, they stated that before one can evaluate the effectiveness of an agent delivered via CED, technical reproducibility must be achieved.  The authors concluded that discouraging results from the 2 randomized phase III studies conducted to date revealed technical shortcomings that need to be addressed to allow CED to fulfill its therapeutic potential; CED holds promise for treating glioblastoma and warrants further pre-clinical and clinical development.

Saito and Tominaga (2017) noted that CED circumvents the BBB by delivering agents directly into the tumor and surrounding parenchyma; CED can achieve large volumes of distribution by continuous positive-pressure infusion.  Although promising as an effective drug delivery method in concept, the administration of therapeutic agents via CED is not without challenges.  Limitations of distribution remain a problem in large brains, such as those of humans.  Accurate and consistent delivery of an agent is another challenge associated with CED.  Similar to the difficulties caused by immunosuppressive environments associated with gliomas, there are several mechanisms that make effective local drug distribution difficult in malignant gliomas.  These investigators discussed methods for local drug application targeting gliomas with special emphasis on CED.  The authors concluded that although early clinical trials have failed to demonstrate the efficacy of CED against gliomas, CED potentially can be a platform for translating the molecular understanding of glioblastomas achieved in the laboratory into effective clinical treatments.   They noted that several clinical studies using CED of chemotherapeutic agents are ongoing; successful delivery of effective agents should prove the efficacy of CED in the near future.

Sasaki and colleagues (2020) examined the efficacy of enhancer of zeste homolog-2 (EZH2) inhibitor by CED against human DIPG xenograft models.  The concentration of EZH2 inhibitor (EPZ-6438) in the brainstem tumor was evaluated by liquid chromatography-mass spectrometry (LC/MS).  These researchers treated mice-bearing human DIPG xenografts with EPZ-6438 using systemic (intra-peritoneal) or CED administration.  Intra-cranial tumor growth was monitored by bioluminescence image, and the therapeutic response was evaluated by animal survival.  LC/MS analysis showed that the concentration of EPZ-6438 in the brainstem tumor was 3.74 % of serum concentration following systemic administration.  CED of EPZ-6438 suppressed tumor growth and significantly extended animal survival when compared to systemic administration of EPZ-6438 (p = 0.0475).  The authors concluded that the findings of this study indicated that CED of an EZH2 inhibitor is a promising strategy to bypass the BBB and to increase the efficacy of an EZH2 inhibitor for the treatment of DIPG.

Bander and associates (2020) reported on the safety and experience in a group of pediatric patients who received sequential CED into the brainstem for the treatment of DIPG.  Patients in this study were enrolled in a single-center, phase-I clinical trial using 124I-8H9 monoclonal antibody (124I-omburtamab) administered by CED.  A retrospective chart and imaging review were used to examine demographic data, CED infusion data, and post-operative neurological and surgical outcomes.  MRI scans were analyzed using iPlan Flow software for volumetric measurements.  Target and catheter coordinates as well as radial, depth, and absolute error in MRI space were calculated with the ClearPoint imaging software.  A total of 7 patients underwent 2 or more sequential CED infusions.  No patients experienced Clinical Terminology Criteria for adverse events (AEs) of grade-3 or greater deficits; 1 patient had a persistent grade-2 cranial nerve deficit after a 2nd infusion.  No patient experienced hemorrhage or stroke post-operatively.  There was a statistically significant decrease in radial error (p = 0.005) and absolute tip error (p = 0.008) for the 2nd infusion compared with the initial infusion.  Sequential infusions did not result in significantly different distribution capacities between the 1st and 2nd infusions (volume of distribution determined by the PET signal/volume of infusion ratio [mean ± SD]: 2.66 ± 0.35 versus 2.42 ± 0.75; p = 0.45).  The authors concluded that the findings of this study showed the ability to safely carry out sequential CED infusions into the pediatric brainstem.  Past treatments did not negatively influence the procedural workflow, technical application of the targeting interface, or distribution capacity.  These researchers stated that this limited experience provided a foundation for using repeat CED for oncological purposes.

In a retrospective review, Szychot and co-workers (2021) described their clinical experience and what they had learned regarding the safety and feasibility of treating DIPG with intermittent CED of carboplatin and sodium valproate to the pons via the Renishaw Drug Delivery System (RDDS).  This study was a review (2017 to 2020) of children with DIPG, who following radiotherapy, received compassionate treatment commencing 3.3 to 10 months post-diagnosis (median of 4.9 months).  They received up to 7 cycles of 3 to 6 weekly pontine infusions of carboplatin (0.12 to 0.18 mg/ml) and sodium valproate (14.4 to 28.8 mg/ml).  A total of 13 children aged 3 to 19 years (mean of 6.9 years) were treated.  There were no surgical complications.  With the exception of infusion channels blocking in 1 device, there were no adverse device effects; 2 patients developed persistent 6th nerve palsies, which led to drug concentration reduction in the combination therapy.  Subsequently infusion/ drug-related toxicities were transient.  Tumor was controlled in pons in 10/13 patients.  Median progression-free survival (PFS) was 13.0 months, while median overall survival (OS) was 15.3 months.  The authors concluded that the use of the RDDS was safe and well-tolerated in all 13 patients, and treatment improved control of pontine disease resulting in longer PFS and OS and merits further evaluation in a clinical trial.

Wang et al (2021) noted that in patients with glioblastoma, resistance to the chemotherapeutic temozolomide (TMZ) limits any survival benefits conferred by the drug.  These researchers showed that CED of nanoparticles containing disulfide bonds (which are cleaved in the reductive environment of the tumor) and encapsulating an oxaliplatin pro-drug and a cationic DNA intercalator inhibited the growth of TMZ-resistant cells from patient-derived xenografts, and hindered the progression of TMZ-resistant human glioblastoma tumors in mice without causing any detectable toxicity.  Genome-wide RNA profiling and metabolomic analyses of a glioma cell line treated with the cationic intercalator or with TMZ showed substantial differences in the signaling and metabolic pathways altered by each drug.  The authors concluded that the findings of this study suggested that the combination of anti-cancer drugs with distinct mechanisms of action with selective drug release and CED may represent a translational strategy for the treatment of TMZ-resistant gliomas.

Sunli et al (2021) stated that in recent years, combination therapy has emerged as the cornerstone of clinical practice in the treatment of GBM; however, their ability to trigger and leverage the body's adaptive immunity has rarely been studied.  Tumor heterogeneity, the presence of the BBB, and an immunosuppressive tumor micro-environment play a crucial role in the 90 % local tumor recurrence post-treatment.  These researchers described an improved combination therapy approach capable of stimulating an immune response that uses light responsive antigen-capturing oxygen generators (LAGs).  The engineered LAGs loaded with a non-genotoxic molecule, Nutlin-3a, and a photosensitizer, protoporphyrin IX, can release the payload on-demand when exposed to light of a specific wavelength.  The in-situ oxygen generation capability of LAGs enables tumor oxygenation enhancement, thereby alleviating the tumor hypoxia and enhancing the efficacy of chemo-photodynamic therapy (PDT).  Furthermore, by modulating the surface properties of LAGs, these researchers demonstrated that the tumor-derived protein antigens released can be captured and retained in-situ, which improved antigen uptake and presentation by the antigen-presenting cells.  Dual drug-loaded LAGs (DD-LAGs) up-regulated the expression of cell surface CD83 maturation and CD86 co-stimulatory markers on monocyte-derived-dendritic cells, suggesting intrinsic immune adjuvancy.  In the presence of three-dimensional (3D) printed hypoxic U87 spheroids (h-U87), DD-LAGs induced cancer cell death, up-regulated IL-1β, and down-regulated IL-10 resulting in CD3+, helper CD4+, and cytotoxic CD8+ proliferation.  Finally, the authors had examined CED as a potential route of administration for DD-LAGs.  These investigators stated that their work presents a novel strategy to induce tumor cell death both during and post-treatment, thereby reducing the possibility of recurrence.

Lambride et al (2022) noted that brain cancer therapy remains a formidable challenge in oncology; and CED is an innovative and promising local drug delivery method for the treatment of brain cancer, overcoming the challenges of the systemic delivery of drugs to the brain.  To improve the understanding regarding the effectiveness of CED and drug transport, these investigators presented an in-silico methodology for brain cancer CED treatment simulation.  To achieve this, a 3D finite element (FE) formulation was used that employed a brain model representation from clinical imaging data and was used to predict the drug deposition in CED regimes.  The model encompasses biofluid dynamics and the transport of drugs in the brain parenchyma.  Drug distribution was studied under various pathophysiological conditions of the tumor, in terms of tumor vessel wall pore size and tumor tissue hydraulic conductivity as well as for drugs of various sizes, spanning from small molecules to nanoparticles.  By means of a parametric study, these researchers reported the impact of the size of the vascular wall pores and that of the therapeutic agent on drug distribution during and after CED.  The authors concluded that the in-silico findings provided useful insights of the spatiotemporal distribution and average drug concentration in the tumor towards an effective treatment of brain cancer. 

These investigators stated that it is important to acknowledge the simplifications and limitations of the current model.  For the sake of simplicity, the drug was considered a spherical particle and the vessel wall openings were modeled as perfect cylindrical pores; thus, the theory for hindered transport of rigid solutes via liquid filled pores could be applied to describe drug transport across the tumor vessel walls.  Furthermore, to simplify the modeling procedure, the flow physics of the catheter domain were disregarded.  This modeling approach was substantially less challenging, as the complexity (in terms of the extra differential equations and the additional model parameters and boundary conditions) and the considerable computational burden (solving the Navier-Stokes equations and the coupling of these with the biphasic FE formulation) were minimized.  The catheter jet flow was modeled by taking appropriate boundary conditions at the interface between the catheter and the (tumor or host) tissue in order to simulate the drug administration during CED.  After CED administration, a zero-flux boundary condition was used on the outlet surface of the catheter, neglecting any drug amount that could be diffused from the catheter to the tumor tissue.  Regarding the model limitations, it incorporates only biomechanical properties of the grey and white matter, while it does not account for other components of the brain, such as the thalamus, the internal capsule, the corpus callosum, the putamen, ventricles, and cavities.  How the predictive results would be affected by the incorporation of the other brain components is not intuitive; therefore, detailed simulations would have to be performed.  In addition, according to the literature, the drug transport efficiency varies greatly in different regions of the brain, since the effective diffusivity in the gray matter is isotropic, whereas white matter diffusion is anisotropic.  Hence, diffusion tensor imaging (DTI) data could be considered to provide estimates of the tissue anisotropy within the entire brain and, therefore, reduce these uncertainties.

Brady et al (2022) stated that limitations have previously existed for the use of brain infusion catheters with extended delivery port designs to achieve larger distribution volumes using CED, due to poor transmittance of materials and uncontrolled backflow.  These researchers examined a novel brain catheter that has been designed to allow for extended delivery and larger distribution volumes with limited backflow of fluid.  It was characterized using a broad range of therapeutic pore sizes both for transmittance across the membranes to address possible occlusion and for distribution in short-term infusion studies, both in-vitro in gels and in-vivo in canines.  Brain catheters with pore sizes of 10, 12, 15, 20 and 30 µm were examined using 3 infusates prepared in 0.9 % sterile saline with diameters approximating 2, 5, and 30 nm, respectively.  Magnevist was chosen as the small molecule infusate to mimic low-molecular weight therapeutics.  Galbumin served as a surrogate for an assortment of proteins used for brain cancer and PD.  Gadoluminate was used to evaluate the distribution of large therapeutics, such as adeno-associated viral particles and synthetic nanoparticles.  The transmittance of the medium and large tracer particles via catheters of different pore size (15, 20 and 30 µm) was measured by MRI and compared with the measured concentration of the control.  Infusions into 0.2 % agarose gels were carried out to examine differences in transmittance and distribution of the small, medium, and large tracer particles via catheters with different pore sizes (10, 12, 15, 20 and 30 µm).  In-vivo infusions were carried out in the canine to examine the ability of the catheter to infuse the small, medium, and large tracer particles into brain parenchyma at high flow rates through catheters with different pore sizes (10, 15, and 20 µm).  Two catheters were inserted stereotactically into the brain for infusion, one per hemisphere, in each animal (n = 6).  The transmittance of Galbumin and Gadoluminate across the catheter membrane surface was 100 % to within the accuracy of the measurements.  There was no evidence of any blockage or retardation of any of the infusates.  Catheter pore size did not appear to significantly affect transmittance or distribution in gels of any of the molecule sizes in the range of catheter pore sizes tested.  There were differences in the distributions between the different tracer molecules: Magnevist produced relatively large distributions, followed by Gadoluminate and Galbumin.  These investigators observed no instances of uncontrolled backflow in a total of 12 in-vivo infusions.  Furthermore, several of the infusions resulted in substantial amounts remaining in tissue.  They expected the in-tissue distributions to be substantially improved in the larger human brain.  The new porous brain catheter performed well in terms of both backflow and intra-parenchymal infusion of molecules of varying size in the canine brain under CED flow conditions.  The authors concluded that overall, the data presented in this report support that the novel porous brain catheter can deliver therapeutics of varying sizes at high infusion rates in the brain parenchyma, and resist backflow that can compromise the effectiveness of CED therapy.  Moreover, these researchers stated that further investigation is needed to characterize the brain catheter, including animal toxicity studies of chronically implanted brain catheters to lay the foundation for its use in the clinic.

Rechberger et al (2023) noted that despite much progress, the prognosis for H3K27-altered diffuse midline glioma (DMG, formerly known as diffuse intrinsic pontine glioma) when located in the brainstem, remains dark and dismal.  Extensive research over the last 10 years has revolutionized the understanding of the molecular basis of DMG, showing potential targetable vulnerabilities for the treatment of this lethal childhood cancer.  However, obstacles to successful clinical implementation of novel therapies remain, including effective delivery across the BBB to the tumor site.  These investigators reviewed relevant literature and clinical trials and discussed direct drug delivery via CED as a promising treatment modality for DMG.  They outlined a comprehensive molecular, pharmacological, and procedural approach that may offer hope for afflicted patients and their families.  The authors concluded that challenges remain in successful drug delivery to DMG.  While CED and other techniques offer a chance to bypass the BBB, the variables influencing successful intra-tumoral targeting are numerous and complex.  These researchers discussed these variables and potential solutions that could lead to the successful clinical implementation of pre-clinically promising therapeutic agents.


References

The above policy is based on the following references:

  1. Anderson RC, 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.
  2. Aparicio-Blanco J, Torres-Suarez AI. Glioblastoma multiforme and lipid nanocapsules: A review. J Biomed Nanotechnol. 2015;11(8):1283-1311.
  3. 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.
  4. Barua NU, Bienemann AS, Hesketh S, et al. Intrastriatal convection-enhanced delivery results in widespread perivascular distribution in a pre-clinical model. Fluids Barriers CNS. 2012;9(1):2.
  5. Barua NU, Gill SS, Love S. Convection-enhanced drug delivery to the brain: Therapeutic potential and neuropathological considerations. Brain Pathol. 2014;24(2):117-127.
  6. Bidros DS, Liu JK, Vogelbaum MA. Future of convection-enhanced delivery in the treatment of brain tumors. Future Oncol. 2010;6(1):117-125.
  7. Bidros DS, Vogelbaum MA. Novel drug delivery strategies in neuro-oncology. Neurotherapeutics. 2009;6(3):539-546.
  8. Brady ML, Grondin R, Zhang Z, et al. In-vitro and in-vivo performance studies of a porous infusion catheter designed for intraparenchymal delivery of therapeutic agents of varying size. J Neurosci Methods. 2022;378:109643.
  9. Brady ML, Raghavan R, Singh D, et al. In vivo performance of a microfabricated catheter for intraparenchymal delivery. J Neurosci Methods. 2014;229:76-83.
  10. Bruce JN, Fine RL, Canoll P, et al. Regression of recurrent malignant gliomas with convection-enhanced delivery of topotecan. Neurosurgery. 2011;69(6):1272-1279; discussion 1279-1280.
  11. Buonerba C, Di Lorenzo G, Marinelli A, et al. A comprehensive outlook on intracerebral therapy of malignant gliomas. Crit Rev Oncol Hematol. 2011;80(1):54-68.
  12. Centers for Medicare and Medicaid Services (CMS), ICD-9-CM Coordination and Maintenance Committee. ICD-9-CM Volume III, Procedures. Agenda. Baltimore, MD: CMS; March 23-24, 2006.
  13. Centers for Medicare and Medicaid Services (CMS). Decision Memo for Blood Brain Barrier Disruption (BBBD) Chemotherapy (CAG-00333N). Medicare Coverage Database. Baltimore, MD: CMS; March 20, 2007.
  14. Fiandaca MS, Forsayeth JR, Dickinson PJ, Bankiewicz KS. Image-guided convection-enhanced delivery platform in the treatment of neurological diseases. Neurotherapeutics. 2008;5(1):123-127.
  15. Garg T, Bhandari S, Rath G, Goyal AK. Current strategies for targeted delivery of bio-active drug molecules in the treatment of brain tumor. J Drug Target. 2015;23(10):865-887.
  16. Hall WA, Sherr GT. Convection-enhanced delivery of targeted toxins for malignant glioma. Expert Opin Drug Deliv. 2006;3(3):371-377.
  17. Han J, Yang L, Puri RK. Analysis of target genes induced by IL-13 cytotoxin in human glioblastoma cells. J Neurooncol. 2005;72(1):35-46.
  18. Institute for Clinical Systems Improvement (ICSI). Blood brain barrier disruption chemotherapy. Technical Assessment Report: 59. Bloomington, MN: ICSI; November 2001.
  19. Jahangiri A, Chin AT, Flanigan PM, et al. Convection-enhanced delivery in glioblastoma: A review of preclinical and clinical studies. J Neurosurg. 2017;126(1):191-200.
  20. Kaal EC, Niel LG, Vecht CJ. Therapeutic management of brain metastases. Lancet Neurol. 2005;4(5):289-298.
  21. Kawakami K, Kawakami M, Puri RK. Nitric oxide accelerates interleukin-13 cytotoxin-mediated regression in head and neck cancer animal model. Clin Cancer Res. 2004;10(15):5264-5270.
  22. Kawakami K, Kioi M, Liu Q, et al. Evidence that IL-13R alpha2 chain in human glioma cells is responsible for the antitumor activity mediated by receptor-directed cytotoxin therapy. J Immunother. 2005;28(3):193-202.
  23. Kawakami K, Nakajima O, Morishita R, Nagai R. Targeted anticancer immunotoxins and cytotoxic agents with direct killing moieties. Sci World J. 2006;6:781-790.
  24. Kim I, Paek S, Nelson BD, et al. Implementation of a chronic unilateral intraparenchymal drug delivery system in a swine model. J Neurosci Methods. 2014;227:29-34.
  25. Kioi M, Husain SR, Croteau D, et al. Convection-enhanced delivery of interleukin-13 receptor-directed cytotoxin for malignant glioma therapy. Technol Cancer Res Treat. 2006;5(3):239-250.
  26. Kunwar S, Chang SM, Prados MD, et al, Safety of intraparenchymal convection-enhanced delivery of cintredekin besudotox in early-phase studies. Neurosurg Focus. 2006;20(4):E15.
  27. Kunwar S. Convection enhanced delivery of IL13-PE38QQR for treatment of recurrent malignant glioma: Presentation of interim findings from ongoing phase 1 studies. Acta Neurochir Suppl. 2003;88:105-111.
  28. Lam MF, Thomas MG, Lind CR. Neurosurgical convection-enhanced delivery of treatments for Parkinson's disease. J Clin Neurosci. 2011;18(9):1163-1167.
  29. Lambride C, Vavourakis V, Stylianopoulos T, et al. Convection-enhanced delivery in silico study for brain cancer treatment. Front Bioeng Biotechnol. 2022;10:867552.
  30. Mendiburu-Eliçabe M, Gil-Ranedo J. Combination therapy of intraperitoneal rapamycin and convection-enhanced delivery of nanoliposomal CPT-11 in rodent orthotopic brain tumor xenografts. Curr Cancer Drug Targets. 2015;15(4):352-362.
  31. Muro K, Das S, Raizer JJ. Convection-enhanced and local delivery of targeted cytotoxins in the treatment of malignant gliomas. Technol Cancer Res Treat. 2006;5(3):201-213.
  32. NeoPharm, Inc. NeoPharm provides update on reimbursement progress for cintredekin besudotox. Unique coding provided for convection enhanced delivery. Press Release. Waukegan, IL: NeoPharm; August 7, 2006.
  33. Patel MM, Patel BM. Crossing the blood-brain barrier: Recent advances in drug delivery to the brain. CNS Drugs. 2017;31(2):109-133. 
  34. Pleticha J, Maus TP, Christner JA, et al. Minimally invasive convection-enhanced delivery of biologics into dorsal root ganglia: Validation in the pig model and prospective modeling in humans. Technical note. J Neurosurg. 2014;121(4):851-858.
  35. Raghavan R, Brady ML, Rodríguez-Ponce MI, et al. Convection-enhanced delivery of therapeutics for brain disease, and its optimization. Neurosurg Focus. 2006;20(4):E12.
  36. Rainov NG, Soling A. Technology evaluation: Cintredekin besudotox, NeoPharm/Nippon. Curr Opin Mol Ther. 2005;7(2):170-181.
  37. Rechberger JS, Power BT, Power EA, et al. H3K27-altered diffuse midline glioma: A paradigm shifting opportunity in direct delivery of targeted therapeutics. Expert Opin Ther Targets. 2023;27(1):9-17.
  38. Rogawski MA. Convection-enhanced delivery in the treatment of epilepsy. Neurotherapeutics. 2009;6(2):344-351.
  39. Saito R, Tominaga T. Convection-enhanced delivery of therapeutics for malignant gliomas. Neurol Med Chir (Tokyo). 2017;57(1):8-16.
  40. Sampson JH, Akabani G, Archer GE, et al. Intracerebral infusion of an EGFR-targeted toxin in recurrent malignant brain tumors. Neuro Oncol. 2008;10(3):320-329.
  41. Sampson JH, Akabani G, Friedman AH, et al. Comparison of intratumoral bolus injection and convection-enhanced delivery of radiolabeled antitenascin-monoclonal antibodies. Neurosurg Focus. 2006;20(4):E14.
  42. Sampson JH, Archer G, Pedain C, et al; PRECISE Trial Investigators. Poor drug distribution as a possible explanation for the results of the PRECISE trial. J Neurosurg. 2010;113(2):301-309.
  43. Sasaki T, Katagi H, Goldman S, et al. Convection-enhanced delivery of enhancer of zeste homolog-2 (EZH2) inhibitor for the treatment of diffuse intrinsic pontine glioma. Neurosurgery. 2020;87(6):E680-E688.
  44. 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.
  45. Shi M, Fortin D, Sanche L, Paquette B. Convection-enhancement delivery of platinum-based drugs and Lipoplatin(TM) to optimize the concomitant effect with radiotherapy in F98 glioma rat model. Invest New Drugs. 2015;33(3):555-563.
  46. Shimamura T, Husain SR, Puri RK. The IL-4 and IL-13 pseudomonas exotoxins: New hope for brain tumor therapy. Neurosurg Focus. 2006;20(4):E11.
  47. Souweidane MM, Occhiogrosso G, Mark EB, Edgar MA. Interstitial infusion of IL13-PE38QQR in the rat brain stem. J Neurooncol. 2004;67(3):287-293.
  48. Stockwell J, Abdi N, Lu X, et al. Novel central nervous system drug delivery systems. Chem Biol Drug Des. 2014;83(5):507-520.
  49. Sugiyama S, Yamashita Y, Kikuchi T, et al. Safety and efficacy of convection-enhanced delivery of ACNU, a hydrophilic nitrosourea, in intracranial brain tumor models. J Neurooncol. 2007;82(1):41-47.
  50. Sunil V, Mozhi A, Zhan W, et al. Convection enhanced delivery of light responsive antigen capturing oxygen generators for chemo-phototherapy triggered adaptive immunity. Biomaterials. 2021;275:120974.
  51. 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.
  52. Wang Y, Jiang Y, Wei D, et al. Nanoparticle-mediated convection-enhanced delivery of a DNA intercalator to gliomas circumvents temozolomide resistance. Nat Biomed Eng. 2021;5(9):1048-1058.
  53. White E, Bienemann A, Taylor H, et al. A phase I trial of carboplatin administered by convection-enhanced delivery to patients with recurrent/progressive glioblastoma multiforme. Contemp Clin Trials. 2012;33(2):320-331.
  54. Xi G, Robinson E, Mainia-Farmell B, et al. Convection-enhanced delivery of nanodiamond drug delivery platforms for intracranial tumor treatment. Nanomedicine. 2014;10(2):381-391.
  55. Yamashita Y, Krauze MT, Kawaguchi T, et al. Convection-enhanced delivery of a topoisomerase I inhibitor (nanoliposomal topotecan) and a topoisomerase II inhibitor (pegylated liposomal doxorubicin) in intracranial brain tumor xenografts. Neuro Oncol. 2007;9(1):20-28.
  56. Zhou Z, Singh R, Souweidane MM, et al. Convection-enhanced delivery for diffuse intrinsic pontine glioma treatment. Curr Neuropharmacol. 2017;15(1):116-128.