Grants

The DIPG / DMG Collaborative has funded $17,296,520 in DIPG/DMG research.

Interested in applying for a grant from the DIPG / DMG Collaborative? Learn more.




Children's Cancer Institute - $99,251

Amount
$99,251.00
Date

September 2020

Discovering New Epigenetic Players in DIPG Oncogenesis

Diffuse intrinsic pontine glioma (DIPG) is the most aggressive brain tumour in children. DIPG accounts for 10-15 percent of all paediatric central nervous system tumours. Every year in US 300 children are diagnosed with DIPG and 300 children will die of this disease. By conducting fundamental research, we will investigate companion proteins, called epigenetic factors, of H3.3K27M, the most prevalent mutation found in DIPG, that contribute to the development of this cancer. Despite the fact that these protein partners are key to the normal functioning of H3.3 and have been link to other cancers, they have never been investigated in the context of DIPG. This study will lead to the identification of new therapeutic targets tailored for DIPG, an urgent need of this disease.

2021 Research Update - The H2A.Z-nuclesome code in mammals: emerging functions

Northwestern Memorial Hospital - $110,000

Amount
$110,000.00
Date

September 2020

Development of Histone Demethylase Inhibitor Against DIPG

For a child is diagnosed with a diffuse intrinsic pontine glioma so called DIPG, the options for treatment are scarce and so are the chances for survival. This aggressive brain tumor generally strikes children who are 6 years old and younger, with most surviving less than two year after diagnosis. DIPG has a specific gene mutation, leads to change a function of DNA binding protein, histone. This gene mutation thought to be a specific therapeutic target that
 could be used to make new effective drugs to help kids with DIPG get better. We recently showed that the drug targeting specific histone enzyme, named GSK-J4, delay tumor growth and increase survival of mice implanted human DIPG cells. Because of its promising anti-tumor activities, GSK-J4 has been used to treat many kinds of tumors in animal models including leukemia, lymphoma, prostate and gastric cancer, and DIPG; however, GSK-J4 is not yet in clinical development for treating patients. The major challenge for GSK-J4 in clinical development is that GSK-J4 is a drug that is rapidly changed to the active drug GSKJ1, which has limited to enter the tumor cells in the brain. We have developed a novel compound that is derived from GSK-J1, named UR-8, as a lead anti-cancer compound through our drug screening. UR-8 has showed selective cytotoxic activity against human DIPG cells and apparently transported to the brain with a useful extent based on its in vivo stability and effectiveness. UR-8 showed greater anti-tumor activity and survival benefit than that obtained by GSK-J4 treatment in human DIPG animal models. Based on the observation of this lead compound, UR-8, we will further develop new drugs which are similar to GSK-J4/J1 to increase the potency, specificity, and brain transport features. We will test cytotoxicity and assess safety and tolerability of new compounds in DIPG mouse models.
Our long-term goal is to use new compounds in combination with radiation which is used routinely for the treatment of DIPG patient. We will treat DIPG mouse models with a new compound alone or in combination with radiation. Our basic drug discovery, in combination with therapeutic testing in animal models, in turn, will therefore provide insights for testing a novel therapeutic approach for treating currently incurable pediatric brain tumors.

The Hospital for Sick Children - $100,000

Amount
$100,000.00
Date

September 2020

Methionne Deptrivation as a Therapeutic Strategy for Diffuse Intrinsic Pontine Glioma

After decades of clinical trials, no effective treatment has yet been found for DIPG. While we now know a lot about the genetics of this disease, this has unfortunately not thus far led to improved survival for children with DIPG. To better understand the disrupted pathways in DIPG we performed an analysis of their proteins and how these differed from normal brain. One of the interesting pathways that came up was the methionine (an essential amino acid) metabolism pathway. We then tested DIPG patient cells and found that they are particularly sensitive to methionine withdrawal. Here we propose to test the effects of methionine deprivation on cell survival, death, and proliferation as well as methionine metabolism of DIPG cells. We will then test the ability of methionine deprivation to prolong survival in mice harboring DIPG either alone, or together with radiation. We will learn why low methionine levels are detrimental to cancer cells and tolerable to normal cells and how this can be utilized therapeutically. Low methionine diets are very tolerable to humans and are similar to a vegan diet thus this method is readily transferable to the clinic without extended drug approval requirement and expense. Methionine dependency has never before been studied in DIPG and represents a novel approach to therapeutically target DIPG.

Hunter Medical Research Institute - $145,640

Amount
$145,640.00
Date

September 2020

Combatt DMG: Combined Anti-tumor Targeting of Diffuse Midline Gliomas

Paediatric high-grade glioma of the brainstem – diffuse midline glioma (DMG), represent the most aggressive primary tumor of the central nervous system (CNS) and are responsible for half of all brain cancer deaths in children. Recent scientific advances provide much needed insight into the biology of the disease – particularly the genetic characteristics of patient biopsies and autopsies, providing clues regarding the gene mutations that influence cancer growth, and the ability of the tumor to continue to grow, or to sidestep, despite drug treatment. However, the specific knowledge of the mechanisms underpinning how the tumor moves and grow among and through normal brain structures, is still unknown.
Our group shows the drug ‘paxalisib’ (which readily crosses the blood brain barrier) potently inhibits ‘PI3K/Akt/mTOR' – a cell communication axis that influences growth, proliferation and that contributes to energy production. Profiling the activity of proteins (phosphoproteomics) in DMG tumor cells following treatment with paxalisib, identified that the tumor amplified communication (Protein kinase C - PKC) pathways that activate migratory and infiltrative growth dissemination (‘diffuse’/’intrinsic’, hallmarks of DMG). These finding led us to test the hypothesis that dual targeting of PI3K/Akt/mTOR and PKC has the potential to improve outcomes for DMG patients by stopping proliferative and infiltrative growth.
For the first time, we have subjected DMG cells to global phosphoproteomic profiling, +/- treatment with paxalisib and rapamycin (mTOR inhibitor), to expose the signalling pathways responsible for continued growth, survival and ultimately the development of treatment resistance. Our efforts sought to identify which of the spectrum of pathways become over-activated in patients following PI3K inhibition, in the hope that these pathways could be simultaneously targeted at diagnosis and/or at disease progression and/or as a drug target when resistance occurs. All DMG cell lines were sensitive to paxalisib (0.197-1.128 IC ); with biochemical investigations confirming potent inhibition of PI3K/Akt signaling. However, PI3K or mTOR inhibition potently activated PKC, and downstream proteins that control the dynamic remodeling of the actin cytoskeleton that influence cell motility, alter cell polarity and drive invasion of the tumor; aiding escape from radiation therapy, and in this case, PI3K and mTOR inhibition.
Targeting the PI3K/Akt/mTOR pathway is an approach that has been trialed in many cancers. The pathway is recognized as one of the most overactive growth, proliferation, cellular metabolism pathways in cancer, however, this strategy has yielded little clinical benefit, as therapy resistance quickly develops. The PI3K/AKT/mTOR pathway also plays a pivotal role in the metabolic and mitogenic (cell division) actions of insulin produced in response to increased blood sugar levels. The catalytic enzyme of the PI3K signalling axis, ‘PIK3CA’, is frequently mutated in DMG (PI3KR1, PTEN, MTORC1, TSC1 also).
This is particularly important in DMG, as corticosteroids prescribed to attempt to control hallmark peritumoural inflammation and hydrocephalus have strong affinity for glucocorticoid receptors (so termed "glucocorticoids") which reduce inflammation, but also impact on glucose homeostasis, increasing blood glucose levels and stimulating appetite.
Recently, blood glucose and insulin levels were monitored in mice treated with several PI3K/Akt inhibitors (paxalisib excluded) currently in human clinical trials and showed PI3K inhibitors elevated blood glucose and insulin levels.
Furthermore, injection of insulin abrogated the antitumor efficacy of PI3K inhibitors in mice harboring PI3K-mutant breast cancers. These results, coupled with our identification of PKC activation in response to PI3K inhibition, provide a
mechanistic link that underpins resistance to PI3K/Akt/mTOR inhibitors in DMG that warrants rigorous investigation.
Our poilot data showed high-level synergistic cell death in 88% of DMG cell lines when treated with PI3K inhibitors in combination with PKC inhibitors, FDA approved to treat other cancers. Assessment of the level of PKC signaling in untreated DMG cell lines, revealed overactivation in 85% of cell lines; with the highest level of activation seen in patient samples harboring H3.3K27M mutations, linked with shortened overall survival compared to DMG patients diagnosed with H3.1K27M mutations or wildtype H3. In hindsight this is unsurprising: these high-grade gliomas, which originate in the pontine region of the brainstem, frequently disseminate into other midline structures (thalamus) and the cerebellum.
This project builds on strong pilot data, showing the important role of proteins that influence dynamic cellular remodeling, driving growth and migration into surrounding and distant tissues in advanced disease. Using complementary molecular (CRISPR-cas9 and shRNA), and pharmacological studies, coupled with sophisticated and
quantitative protein sequencing technologies, cell biology techniques and pre-clinical models, we will assess the functional role of PKC and activated actin cytoskeleton remodeling proteins, that influence cell motility, cell polarity, to drive invasion of the tumor; to aid in escape from radiation and emerging targeted therapies. This project will determine whether simultaneous targeting of growth/proliferation/metabolic pathways in combination with cell motility pathways is a treatment paradigm that will improve outcomes for patients.

International DIPG/DMG Registry And Repository - $862,671

Amount
$862,671.00
Date

May 2020

Diffuse intrinsic pontine gliomas (DIPG) are the most common brainstem tumors in children, representing approximately 75-80% of all pediatric brainstem tumors. Approximately 200-300 patients are diagnosed with DIPG North America and a similar number in Europe.  DIPG accounts for 10-15% of all new pediatric brain tumor diagnoses and is the leading cause of brain tumor-related death in children. The median age at diagnosis is 6 to 7 years with a median survival of less than 1 year. Although radiotherapy does improve neurological function and survival by 2-3 months, no effective chemotherapeutic regimens are currently available. The  International Diffuse Intrinsic Pontine Glioma/Diffuse Midline Glioma Registry (IDIPG/DMGR) represents  the largest and most comprehensive collection of linked clinical, radiologic, pathologic and gemomics data  from a diverse cohort of DIPG patients available to researchers throughout the world.  With the generous support of a coalition of pediatric brain tumor foundations forming the DIPG Collaborative, and collaborations with 113 academic medical centers in 15 countries, the IDIPG/DMGR continues to expand. From April 2012 to December 2019, 1086 patients diagnosed with DIPG have been enrolled.  The radiology repository contains 5405 studies from over 700 patients. Images from 589 patients have been centrally reviewed. The pathology repository contains 3562 specimens on 125 patients. Sixty-two autopsies have been coordinated by the IDIPGR.  Fresh DIPG tissue from 20 autopsies has been sent to investigators to develop primary cell cultures. Five   primary cell lines have been established. 199 genomic data sets are available including whole genome, whole exome, and/or RNA seq, and methylation.  Investigators from around the world are currently conducting 20 projects using data/tissue from the DIPG/DMG Registry.  Six additional studies have been published. In 2019, the steering committee of the IDIPGR approved the expansion of the Registry to include patients with diffuse midline gliomas. The first 11 patients with DMG have now been enrolled in the IDIPG/DMG Registry.

Sydney Children's Hospital - $151,468

Amount
$151,468.00
Date

December 2019

Developing New Epigenetic Combination Treatments Against DIPG.

Diffuse intrinsic pontine glioma (DIPG) is the most aggressive of all childhood cancers. Standard treatment with radiotherapy is only palliative and single drug chemotherapy is ineffective. We have found that DIPG cells are sensitive to a novel anticancer treatment. CBL0137 is a novel class of anticancer agents that can inhibit the action of a key protein complex called Facilitates Chromatin Transcription (FACT). The FACT complex controls multiple cancer –associated pathways that can lead to aberrant tumor growth. We have found that CBL0137 can inhibit further the growth of DIPG tumors when combined with other anticancer drugs such as panobinostat and JQ1. Furthermore preliminary testing has shown that CBL0137 can also be combined with other chemotherapeutic agents such as olaparib and ACT001. Olaparib is a clinically approved agent for ovarian and breast cancers where as ACT001 is currently in Phase 1 testing in pediatric patients with DIPG. In this project, we aim to develop these new combination therapy strategies in preclinical models of DIPG and obtain the necessary quantum of data to begin the transition of CBL0137 therapy from the bench to the bedside to directly benefit children with this devastating and currently incurable tumor.

University of Sydney - $100,000

Amount
$100,000.00
Date

November 2019

Targeting hypoxia and mitochondrial metabolism with repurposing drugs as an approach of radiosensitization for diffuse intrinsic pontine gliomas.

Diffuse intrinsic pontine glioma (DIPG) is a rare and incurable brain tumor that arises in the brainstem of children predominantly between the ages of 6 and 9. Unlike many brain tumors, DIPGs cannot be removed through surgery due to its sensitive location. The standard of care today remains radiation therapy (RT) alone. Unfortunately, almost all DIPGs recur locally within 12 months secondary to radioresistance. Therefore, it is important to understand the mechanisms of radioresistance, as this may be used to improve the radiosensitivity and offer a pathway to the development of novel therapies for this deadly brain tumor. Hypoxia, a condition in which the body is deprived of adequate oxygen supply at the tissue level, is a common microenvironmental feature of all solid tumors, playing a vital role in radioresistance. Recent reports showed that DIPGs are exposed to a hypoxic microenvironment, suggesting targeting of hypoxia may be effective to improve their radiosensitivity. My preliminary data have shown that the radiosensitivity of DIPG cells was significantly improved when treated with biguanide, a class of diabetes drug that can reduce hypoxia, and this radiosensitising effect was further improved when a second drug was combined to further modulate sugar metabolism. These findings could potentially form the basis for pharmaceutically targeting hypoxia and tumor metabolism as a new radiosensitising treatment for incurable DIPG. Children currently diagnosed with DIPG have no hope of cure and are offered palliative treatment only. RT is the only effective treatment for DIPG to date although it only provides relief of tumor-related symptoms in roughly 70% patients. However, all DIPGs recur locally secondary to radioresistance, thus improving the effect of RT remains the most promising avenue to better outcomes in DIPG patients. Cells under hypoxia, a condition where the tissues do not have enough oxygen supply, are 2-3 times more resistant to RT than cells that are well oxygenated at the time of irradiation [1]. A recent study reported that DIPG cells are exposed to a hypoxic microenvironment, suggesting these cells are intrinsically resistant to RT [2]. These findings also suggest that targeting hypoxia may be an effective strategy to overcome the radioresistance of DIPG cells. Biguanides (metformin/phenformin) are a class of drugs that are currently used in clinic for the treatment of Type II diabetes. Apart from lowering blood sugar level, biguanides can also reduce the oxygen consumption of cells by targeting mitochondria, the rod-shaped organelles that can be considered the power generators of cells. By lowering tumor demand for oxygen, the hypoxic condition can thus be lessened. In our pilot studies, we have observed that metformin largely improved the efficacy of RT in a mouse model carrying DIPG cells in their brains. This exciting result led us to further optimize this strategy such that a better efficacy can be achieved. Given metformin is reliant on certain transporters to enter tumor cells [3], we proposed to use a similar drug phenformin as it does not rely on those transporters to enter cells [4], thereby allowing higher drug concentration in tumor cells. Strikingly, phenformin is ~30 times stronger than metformin to slow the growth of DIPG cells. Phenformin reduced oxygen consumption rate which in turn increased lactic acid production. Given high level of lactic acid is the primary adverse effect of phenformin and strongly correlates with poor clinical outcome, a strategy is thus needed to counteract this unfavorable side effect. Dichloroacetate (DCA), a compound that lowers blood lactic acid levels, was selected to offset this side effect, not only because it is an orphan drug to treat high level of lactic acid, it is also well tolerated by children [5]. When DCA was combined with phenformin, the high level of lactic acid production was largely blocked. Surprisingly, the combination also led to a much stronger cell killing effect by depriving tumor cells of energy supply and damaging their DNA. Moreover, the combination also reduced the level of two master regulators that collaboratively enhance the cancer cell growth/survival and metabolic needs through increased uptake of sugar and contribute significantly to radioresistance. When RT was combined with phenformin and DCA, the most effective activity was observed, with the triple combination leading to the lowest number of surviving DIPG cells. More importantly, both phenformin and DCA can readily cross blood-brain-barrier (BBB), have long been used in clinics, and are very well tolerated by young children. Therefore, they are the drugs with the most amount of testing and closest to being tested in clinical trials. Given the results generated from this project will be rapidly translated to clinic and benefit children with DIPG, the anticipated impact of this research project is of great significance: it may lead to a change in treatment regimen resulting in longer survival rates for the pediatric patients with newly diagnosed DIPG.

University of Michigan Hospitals - $157,856

Amount
$157,856.00
Date

November 2019

Therapeutic reversal of pre-natal pontine ID1 signaling in DIPG

Diffuse intrinsic pontine glioma (DIPG) is a lethal pediatric brain tumor wand few children with this diagnosis survive up to two years. Even with the advent of precision-based medicine, experimental therapies have yet to show benefit beyond standard radiation, highlighting the dire need to identify and investigate novel genetic therapeutic targets in DIPG. DIPG researchers now know that the patterns of mutations driving the growth of DIPG are unique, when compared to other childhood tumors and adult high-grade gliomas.  As many as 85% of DIPGs harbor mutations in histone H3 variant H3.3, coded for by the H3F3A gene – and our work and others have shown that this mutation results in critical changes in tumor biology.  It is therefore necessary to consider H3.3 mutational status when investigating novel therapeutic targets and pathways for DIPG.

Recent work by our collaborative team has demonstrated a critical genetic target in DIPG, inhibitor of DNA binding 1 (ID1). Inhibitor of DNA binding (ID) proteins are key regulators of pre-natal tissue growth and its regulation has been tied to the pathogenesis of many diseases including DIPG. Our lab has exciting preliminary data that DIPG tumor cells re-activate ID1 signaling that was used pre-natally to build the normal pons, resulting in many of the most aggressive features in DIPG.  We have also found that cannabidiol (CBD), the non-toxic and non-psychoactive member of the endocannabinoid family found in Cannabis Sativa, can reduce ID1 levels and the viability of DIPG tumor cells in vitro.  In order to define ID1 as a therapeutic target in a clinically relevant manner, it is important to establish whether ID1 can be effectively targeted in mouse models of DIPG and to understand how ID1 signaling may be regulated in different H3.3 mutational subgroups.

The experiments outlined in this protocol will answer meaningful questions about the mechanism underlying ID1-driven DIPG invasion and potentially open novel therapeutic avenues. In particular, the possible use of CBD therapy to target the ID1 pathway in DIPG is a clinically relevant but scientifically unexplored question in DIPG.

Dayton Children's Hospital - $69,600

Amount
$69,600.00
Date

November 2019

Responses of distinct cell populations to PDGFRA inhibitors in diffuse intrinsic pontine glioma.

No cure has been found for diffuse intrinsic pontine glioma (DIPG), after more than 250 clinical trials.  Evidence for excessive signaling by platelet-derived growth factor receptor alpha (PDGFRA) is commonly found in biopsies of DIPG, but attempts to inhibit this protein has failed as a clinical strategy.  Based on recently published studies evaluating the RNA of individual cells, we are testing the hypothesis that only a fraction of DIPG cells are sensitive to treatments that inhibit PDGFRA signaling.  Using a technique that evaluates protein levels in single cells, this will be the first attempt to validate the previously published RNA data suggesting that multiple cell types are present within the tumor, and correlate the patterns with drug sensitivity.  With the long-term goal of deriving rational therapeutic targets from biopsy tissue, this project could improve patient selection for PDGFRA inhibition by characterizing its effects at the level of individual cells and across the different cell types within DIPG.  

Institute of Cancer Research - $121,863

Amount
$121,863.00
Date

November 2019

Targeting Top3A-Amplified DIPG cells by Sirtuin inhibition.

Diffuse intrinsic pontine glioma (DIPG) is a malignant brainstem tumour arising in children representing a major unmet clinical need, with a 2-year survival rate close to zero. With chemotherapy ineffective and surgical intervention not possible, new therapeutic approaches are urgently required based upon the unique biological mechanisms driving DIPG tumorigenesis. We recently discovered amplification of the gene TOP3A to play an important role in a proportion of DIPGs. Critically, cells driven by this genetic alteration to be exquisitely sensitive to inhibitors of sirtuin, which appears to link TOP3A to DNA repair. We plan to explore this in more detail by turning off, and/or turning on the gene in DIPG cells, in order to better understand it’s precise function. We will also screen a range of drugs known to target sirtuins, in order to determine which may be most useful in the clinic, both in cells grown in the laboratory as well as using patient-derived mouse models. We will further look for possible combinations with sirtuin inhibitors in order to further enhance the potential for rapid translation.  

Telethon Kids Institute - $49,512

Amount
$49,512.00
Date

August 2019

Development of a new and effective therapy against Diffuse Intrinsic Pontine Glioma.

Diffuse intrinsic pontine glioma (DIPG) is a rare paediatric brain cancer that is almost universally fatal. Surgery is generally too risky and standard chemotherapy is ineffective. Radiation is the only treatment currently available with proven, yet temporary, benefits. There is no cure for DIPG. Targeted therapy uses drugs aimed at specific molecules involved in tumour growth, preventing the growth and spread of cancer cells. This approach has proven effective against several types of cancers. However, no effective targeted therapy exists against DIPG. In previous experiments, targeted therapy has been used to “switch off” a factor that is crucial for DIPG cells to grow. Surprisingly, following treatment with targeted therapy, DIPG cells not only survive but grow and thrive. A potential factor explaining this conundrum is the plasticity of brain cells. Plasticity allows brain cells to adapt to different conditions in their environment. Just like normal brain cells, DIPG cells also exhibit plasticity, and may be using it to evade drug treatments. Our plan is to use drugs that specifically inhibit DIPG’s plasticity, in combination with targeted therapies already in use for cancer treatment. This approach should enhance the anti-cancer efficacy of targeted therapies, leading to improved survival of DIPG patients. 

Our study will first be conducted in-vitro, using DIPG cells from patients, and then in-vivo, using mice as model organisms. The drugs we plan to use will target ion channels, which are pore-forming proteins found in the cell’s membrane, which control the flow of small charged molecules (like potassium) in and out of the cell. These flows of ions can rapidly change the internal wiring of a cell. For this reason, ion channels have been associated with brain plasticity, and DIPG cells are known to make extra amounts of ion channels. We will employ state-of-the-art techniques to study ion channels in a group of cancer cells obtained from DIPG patients. The techniques for studying ion channels are well established but have not been used by brain cancer researchers. Our first goal is to identify which sets of ion channels are present on the surface of DIPG cells.

Once we have identified the ion channels present in DIPG cells, we will test drugs specifically designed to block these ion channels. Importantly, drugs that block ion channels are already approved for use in the treatment of other neurological conditions such as epilepsy. Hence, these drugs have a better chance to be rapidly trialled in patients. Overall, our work aims to find new and effective treatment options DIPG patients who currently have a very poor life expectancy.

Sydney Children’s Hospital - $175,089

Amount
$175,089.00
Date

November 2018

Polyamine Pathway Metabolism as a Novel Therapeutic Option for Diffuse Intrinsic Pontine Glioma

Diffuse intrinsic pontine glioma (DIPG) is the most aggressive of all childhood cancers. Standard treatment with radiotherapy is only palliative and chemotherapy has been found ineffective. Polyamines are organic compounds essential for key functions of living cells. They can be made by human cells but also generated from intestinal microorganisms and taken from dietary sources. Polyamines are produced at higher levels in cancer cells and contribute thus to aberrant growth of multiple adult and childhood tumours. Our preliminary experiments have shown that polyamines increase the growth of DIPG tumour cells. We have tested a specific combination of drugs that inhibit the production and cellular uptake of polyamines and found this to be highly active against DIPG cells but not in healthy cells. Furthermore, we have tested this combination in two separate orthotopic animal models of DIPG, which recapitulate the human disease, and found that the survival of these animals was significantly enhanced and tumour growth was reduced. This combination was also found to significantly reduce the growth of DIPG cells together with panobinostat, a targeted drug that is currently in a clinical trial for DIPG patients. This project will determine in animal models of DIPG whether polyamine inhibition therapy can enhance the effects of radiotherapy, which is the current standard treatment for DIPG patients. Furthermore, we intend to evaluate the combination of polyamine inhibitors together with panonbinostat, in animal models of DIPG.  Finally we also aim to determine the therapeutic efficacy of polyamine inhibition in other paediatric brain tumor animal models. Positive results from this study will provide the preclinical evidence required to urgently translate these novel discoveries to clinical trial to directly benefit children with DIPG and other aggressive brain tumours.

Cincinnati Children's Hospital - $540,742

Amount
$540,742.00
Date

November 2018

Expansion of the International DIPG Registry.

Diffuse intrinsic pontine gliomas (DIPG) are the most common brainstem tumors in children, representing approximately 75-80% of all pediatric brainstem tumors. 1 Approximately 200-300 patients are diagnosed with DIPG in all of North America and Europe.  1,2 DIPG  accounts for 10-15% of all new pediatric brain tumor diagnoses and is the leading cause of brain tumor-related death in children.The median age at diagnosis is 6 to 7 years 2-4 and  prognosis for patients with DIPGs remains dismal with a median survival of less than 1 year. Although radiotherapy does improve neurological function and survival by 2-3 months, no effective chemotherapeutic regimens are currently available.1,2 Achieving cure for all children with DIPG remains a major goal of pediatric neuro-oncology. In this application, we propose the expansion of the International Diffuse Intrinsic Pontine Glioma Registry which is now the largest and most comprehensive collection of data including clinical, radiologic and pathologic data linked to a bioinformatics repository of molecular data from a diverse cohort of DIPG patients available to researchers throughout the world.  With the generous support of a coalition of pediatric brain tumor foundations from the DIPG Collaborative, and collaborations with 106 academic medical centers in the US, Canada, Australia, New Zealand, Egypt, Brazil, Chile, China, Argentina, India, Japan, Saudi Arabia, United Arab Emirates, and Lebanon, the Registry is growing exponentially. From April 2012 to August 2018, 885 patients diagnosed with DIPG have been enrolled from 106 collaborating institutions. The specific aims include: 1) To continue recruitment of patients diagnosed with DIPG in the International DIPG Registry to greater than 1,000 patients and further expand internationally; 2) To provide a repository of integrated data set compromised of clinical, pathologic, radiologic and molecular features to the clinical research community for promotion of hypothesis generation and analysis and maintain follow up on all cases; 3) To facilitate conduct of autopsies and sharing of fresh tissue for establishment of in vitro and in vivo models by investigators to be shared with investigators around the world; 4) To expand the bioinformatics repository of existing molecular data on DIPGs that can be linked to patient information in the registry through the Links and Viva platform to include prospective data; 5) To develop a program focused on quality of life through a longitudinal study where patient and/or parent proxy-dyads who enroll on the International DIPG Registry will complete Health related quality of life measures at specified time intervals and aid in supportive management; 6) To broaden collaborations among investigators for hypothesis-driven research studies through the registry that will ultimately lead to better classification and more effective treatment of patients with DIPG. In the next year, the International DIPG Registry investigators will promote robust collaborative research projects on all aspects of DIPG, and will continue to make the International DIPG Registry data available to external investigators after review of the proposed research by Scientific Advisory Committee (SAC). Our long-term goal is to expand on the highly collaborative, international, hypothesis-driven research infrastructure to continue to support a wide spectrum of interdisciplinary and translational projects in DIPGs for all investigators. The data collected form a research continuum from basic biology to clinical practice that will ultimately address our primary goals of a) understanding the biology of DIPGs, b) developing more effective therapies and c) developing innovative approaches to diagnosis, response assessment and multidisciplinary treatment and follow-up that will improve patient outcome in addition to maximizing quality of life.

Bambino Gesú Children's Hospital - $99,382

Amount
$99,382.00
Date

November 2018

Investigating the Role of DIPG-Derived Exosomes in Tumor Growth and Invasion.

Diffuse Intrinsic Pontine Glioma (DIPG) is a highly aggressive and infiltrating brain tumor affecting children for which there is no effective treatment.

Sequencing data have revealed  the existence of well defined subgroups of tumors based on age of onset, specific location in the brain, mutations and clinical outcome. Those tumous are characterized by specific recurrent mutations in histone genes which encodes for H3.3 and H3.1 and another mutation in a bone-related gene called ACVR1 which is largely found mutated with the histone H3.1.

Moreover, the existence of heterogeneity among the DIPG cells composing the tumor mass is increasingly evident. Our recent data suggest the presence of a “network” of distinct subpopulations that taken individually differ in their invasion and migration properties, that interact with each other and with the surrounding microenvironment to promote tumorigenesis and the progression of the tumor.

In this context, a crucial role in the communication between DIPG cells and the microenvironment may be represented by exosomes, cellular microvescicles containing RNA, DNA and proteins.

We hypothesise that the exosomes derived from DIPG cells could mediate the intercellular communication and that they could have a specific "oncogenic signature" that is specific to different molecular subgroup, and therefore could be used as an important diagnostic and prognostic marker.

To define the exosome signature of DIPG, we looked at the proteins enclosed in the exosomes and through which cells could exchange signals.

In particular, we used a panel of patient primary-primary cell lines characterized by the DIPG driver mutations (H3.3 K27M, H3.3 K27M/ACVR1 and H3.1 K27M/ACVR1) cultured in stem cell-like conditions, such as neurospheres (NS) and/or adherent to laminin (L).

Moreover, to better derive specific DIPG oncogenic exosomal signature, we used as a comparison pediatric glioblastoma (pGBM), which arise in different locations but maybe mutated in the same histone genes as DIPG.

The analysis of exosomal protein content showed that DIPG and pGBM carry unique proteins in the exosomes, highlighting the differences between the two tumor types, differences that were clearly shown also for the two DIPG subgroups H3.3 and H3.1

Focus of our study was also another molecule transported in the exosome: the micro-RNAs (miRs). Those are small RNA fragments, that may be exchanged among tumor cells and that we also considered for the identification of exosomal oncogenic signature of DIPG.

In this context, we analyzed the miRNA isolated from different patient primary-derived cell lines including different mutation and location subgroups.  Our data showed that H3.1 K27M/ACVR1 DIPG subgroup was strongly different from H3.3 K27M DIPG and pGBM, concluding that the two main histone H3 gene variants leads to distinct oncogenic programs which could be exploited to develop distinct potential therapeutic strategies.

Moreover, since it has been demonstrated that DIPG and pGBM are heterogeneous tumors characterized by the existence of different cell types that co-operate and communicate one to each other, we also isolated single cells from DIPG and pGBM primary cell lines which were later exploited to give raise to different subpopulations and studied for the exosome.

The single cell derived subpopulations were characterized for their morphology, migration and invasion properties and they were different one from each other underlying the heterogeneous nature of DIPG and pGBM tumor masses. Moreover, we also analyzed the exosome communication between tumor cells, confirming that exomes are an important key of signals exchange.

Having consolidated the role of exosomes in DIPG and pGBM cellular connections and having highlighted some important proteins as well as miRNAs, that could be considered as “oncogenic signature” of DIPG and pGBM, our further aim is to investigate their presence in the exosomes from the peripheral blood of DIPG cancer patients.

This will lead to the identification of new diagnostic markers and prognostic strategies for DIPG and pGBM tumors.

Ann and Robert H. Lurie Children's Hospital of Chicago - $35,000

Amount
$35,000.00
Date

November 2018

Credentialing an Improved DIPG Mouse Model.

DIPG is a rare type of childhood brain cancer that is currently incurable. One barrier for progress against DIPG is the development of predictive models, i.e. models in which observations regarding the evaluation of new therapies will accurately predict observations in clinical trials for children with DIPG.   Models are important for a rare disease like DIPG as it would take a very long time to test every new potential therapy in a clinical trial for children with DIPG. While models will always be imperfect, developing improved models can accelerate the development of effective treatments for children with DIPG.  Studies using human DIPG tissue as well as postmortem human pons tissue strongly suggest that a protein called Olig2 is expressed in the original cell that acquires genetic alterations and gives rise to DIPG. Therefore, developing a mouse model that is initiated in Olig2-expressing neonatal brainstem progenitors and harbors genetic alterations commonly seen in DIPG may yield a more predictive model.  Here we propose to characterize an improved DIPG model that arises in Olig2- progenitors and to evaluate whether H3.3K27M, a genetic event present in the majority of DIPGs, influences DIPG tumor cells’ response to radiation, a treatment that children with DIPG receive.