doctor reviewing an x-ray for signs of a brain tumor

Brain Tumor - Glioblastoma

Brain Tumor - Glioblastoma

Last Section Update: 01/2022

Contributor(s): Tina Kaczor, ND/FABNO; Stephen Tapanes, PhD

1 Introduction

Summary and Quick Facts for Glioblastoma

  • Glioblastoma accounts for roughly 16% of all primary brain and central nervous system tumors and about half of all gliomas. There are estimated to be about 13,000 new glioblastoma cases in the United States in 2020.
  • This protocol aims to empower people affected by glioblastoma with knowledge about the disease and how it is typically managed, as well as emerging treatment strategies potentially accessible through clinical trials. The protocol will also present evidence for the potential complementary role of dietary and integrative interventions in glioblastoma management.
  • Surgery, radiotherapy, and chemotherapy are currently used to treat glioblastoma, but are far from ideal interventions, as they can cause side effects and have limited efficacy. Good nutrition can help patients manage the side effects of cancer treatment, maintain energy, avoid infection, and even help fight the disease.

Glioblastoma is an aggressive type of brain tumor.1,2 Surgery, radiotherapy, and chemotherapy are currently used to treat glioblastoma. Although understanding of glioblastoma has improved in recent years, standard care remains limited in its ability to improve overall survival time.1,3-6

Recent prominent cases of glioblastoma—including Senators John McCain and Edward Kennedy—have helped raise awareness about this harrowing disease,7 and researchers are beginning to uncover promising novel therapies.5,8 In recent years, tremendous progress has been made toward developing better treatments for glioblastoma.1

Emerging evidence has identified a potential relationship between a virus called cytomegalovirus (CMV) and the development of glioblastoma.9,10 Also, a groundbreaking study published in 2013 in the New England Journal of Medicine showed the antiviral drug valganciclovir (Valcyte) improved survival in some glioblastoma patients,11 and follow-up studies published in 2020 have further extended these promising findings.12,13 Pioneering work at Duke University using a bioengineered poliovirus produced remarkable response rates in glioblastoma patients.14,15 Evidence of the effects of some off-label drugs in glioblastoma has been encouraging as well.16 For instance, drugs such as metformin17 and cimetidine18 have shown promise in laboratory studies. Also, integrative, natural interventions, such as vitamin D, resveratrol, curcumin, and melatonin are being actively explored, with intriguing preliminary results.19-22

This protocol aims to empower people affected by glioblastoma with knowledge about the disease and how it is typically managed, as well as emerging treatment strategies potentially accessible through clinical trials. This protocol will also present evidence for the potential complementary role of dietary and integrative interventions in glioblastoma management.

This protocol should be consulted along with other relevant protocols, including:

2 Background

There are two main categories of brain cancers: primary cancers, which originate in the brain, and metastatic cancers, which originate elsewhere in the body and spread to the brain. Primary brain cancers may affect people of all ages, although they occur most frequently in children and older adults.23 This protocol focuses on primary brain cancers and glioblastoma in particular.

Primary brain cancers are usually named after the type of brain cells from which the tumor arises.24 Gliomas have been thought to be derived from glial cells in the brain.25 In 2018, researchers determined that glioblastoma arises from special niches of cells in the brain. This niche is called the subventricular zone and it contains neuronal stem cells. It is aberrations in these stem cells that give rise to glioblastoma.26 This shift in our understanding to a more precise origin of glioblastoma should change our approach to treating this disease in the future.27

Primary brain tumors are given a tumor grade based on how abnormal the tumor cells look when viewed under a microscope.25 For cancers in general, the tumor grade provides some information on how quickly a tumor is likely to grow and spread to other tissues. Grade I tumor cells largely resemble normal cells and are referred to as “well-differentiated.” Glioblastoma is a grade IV glioma. The tumor cells do not look like normal cells and are referred to as “undifferentiated.” Glioblastomas tend to grow rapidly and spread into neighboring brain tissues faster than lower-grade tumors. Unlike many other types of aggressive cancers, however, glioblastoma does not usually spread to other organs outside the central nervous system (brain and spinal cord) except in rare cases.28-30

Glioblastoma accounts for roughly 16% of all primary brain and central nervous system tumors and nearly half of all gliomas.31-33 It is estimated that approximately 13,000 Americans will be diagnosed with glioblastoma in 2020.34

3 Risk Factors

Glioblastoma does not have a single definitive cause, but several risk factors for developing glioblastoma have been identified.35,36

Gender

Men are about 50% more likely to develop glioblastoma than women.31,37,38 Also, a woman’s risk goes up after menopause.38 This finding, along with evidence that some gliomas express estrogen receptors, has led to the suggestion that hormones may play a role in the disease.39-41 However, much more research in this area is needed.38,41

Age

The chances of developing glioblastoma increase with age and peak at age 75 to 84 years.42 Because the average lifespan of people in industrialized countries continues to increase, the median age when glioblastoma is diagnosed has risen to 64 years.33,42

Heritage and Genetics

Glioblastoma is about twice as common in people with European-American ancestry than African-American ancestry.33,38 Also, an increased risk of glioblastoma can be inherited within families. About 10 genetic mutations that increase risk of developing glioma have been identified, but most of them confer a relatively small increase in risk.43 In approximately 5% of cases, glioblastoma may also result from genetic diseases such as tuberous sclerosis, Turcot syndrome, multiple endocrine neoplasia type IIA, and neurofibromatosis type I.38,43

Radiation Exposure

People who have been treated with radiation for medical conditions affecting their ears or skin have an increased risk of developing brain tumors.44,45 In addition, radiation to the head for childhood cancers is also a risk factor for brain cancer development later in life.2,38,46 Some limited evidence suggests repeated CT scans of the head and neck region may increase glioma risk in some patients, although these findings have not been firmly established.47

Body Composition

Greater height has been associated with increased glioma and glioblastoma risk.48,49 One study found that men over 190 centimeters (about six feet three inches) were about twice as likely to develop glioblastoma as men between 170 and 174 centimeters.49 Interestingly, additional data suggest people who finished growing at a later age were more likely to develop gliomas.50 Higher Body Mass Index (BMI) is a risk factor for various types of cancers, including glioblastoma.51-53

Non-Ionizing Electromagnetic Radiation Exposure

Between the mid-1990s and early 2000s, the use of mobile and cordless phones increased rapidly.54 These devices emit electromagnetic radiation from their antennas. Laboratory studies demonstrated that brain cells can be affected by electromagnetic fields.55,56 Whether mobile phone use is related to the development of brain tumors has been the subject of much debate.38

In 2011, the World Health Organization International Agency for Research on Cancer warned that the electromagnetic fields generated by mobile phones and other devices that emit similar non-ionizing electromagnetic radiation are “possibly carcinogenic to humans.”54,57,58 This decision was based on data collected from human case-control studies.54 A 2017 review and meta-analysis found that long-term mobile phone use (10 years or more) significantly increased the risk of glioma, but also emphasized that the available evidence is of low quality and more original research is needed before a better conclusion can be drawn.59 Non-ionizing radiation emitted by cells phones does not damage DNA directly, but researchers have proposed several other mechanisms by which cell phone radiofrequency waves may promote cancer.60,61 More research is needed to clarify the relationship, if any, between cell phone use and brain cancers.

Cytomegalovirus

Emerging evidence has explored whether a virus called cytomegalovirus (CMV) may be related to the development of glioblastoma.9,10 Over half of adults in the United States have been exposed to CMV, but relatively few have an active viral infection.62 A study published in the New England Journal of Medicine described several important findings regarding the relationship between CMV and glioblastoma.63 Of the more than 250 glioblastoma patients, the authors detected the presence of CMV in all but one of the participants. Moreover, patients with lower numbers of virus-infected tumor cells survived 33 months on average, while those with higher numbers survived only 13 months. The authors speculated that CMV infection accelerated tumor progression.63,64 Studies to validate this research have had mixed results, and researchers continue to study whether CMV has a role in the development of glioblastoma or whether it can affect the course of the disease.65,66

4 Signs and Symptoms

Signs and symptoms of brain tumors depend on the size of the tumor and its location within the brain. Headaches are often an initial symptom caused by the pressure placed on the inside of the skull or on the brain's ventricular system. Seizures occur in about one-quarter of patients with newly diagnosed glioblastoma and are usually controlled with anticonvulsant drugs throughout the course of the disease.2,67-69 Symptoms affecting cognitive function can be rapid, including memory, balance, language and/or personality changes.

Tumors in some parts of the brain may cause weakness or numbness in the arms, legs, or face; loss of vision; or changes in speech. More subtle symptoms, such as cognitive dysfunction, mood disorders, personality changes, fatigue, and mild memory problems may also arise in patients with larger tumors located in the frontal or temporal lobes, or in the corpus callosum, a structure that connects the two hemispheres of the brain.2,67-70

5 Diagnosis

Imaging

Magnetic resonance imaging (MRI) is the gold standard non-invasive imaging approach to test whether someone has a brain tumor.38,67,71 This test uses a magnetic field and radio waves to generate images of the brain. It can not only find tumors but also provide information that helps guide treatment decisions.70,72 Some imaging tests use a dye called gadolinium, which is injected into a patient's vein. This dye provides what is referred to as “contrast” and helps distinguish tumor tissue from normal tissue. Patients with suspected glioblastoma may have MRI scans both with and without contrast.37

Other types of imaging tests may be used to complement MRI findings. One of these tests, called MR perfusion, can measure blood flow in tumors and requires a contrast dye.70,73 Another imaging test called MR spectroscopy couples MRI scans with tests to determine what kinds of chemicals are present in the tumor and in the normal surrounding tissues.70,72

A computed tomography (CT) scan is an imaging test usually reserved for patients who cannot undergo an MRI for various reasons.25 For example, patients with pacemakers, or those with certain kinds of cardiac monitors or surgical clips are not candidates for MRI because of the magnetic fields that MRI requires.70 CT scans use X-rays instead of magnetic fields and are also done with and without contrast to provide detailed pictures of the brain.

Additional, more sophisticated imaging tests may be needed to distinguish glioblastomas from cancers that spread from other body parts to the brain.74-76

Biopsy

Although MRI and CT scans can provide valuable information regarding the features of glioblastoma, actual brain tissue is required for a definitive diagnosis.25 During a procedure called a biopsy, a small sample of the brain tumor tissue is removed for further analyses under a microscope.25,70 The tumor tissue from a biopsy is analyzed by a doctor called a pathologist. In addition to determining whether the tumor is glioblastoma, the pathologist may also request a molecular analysis of the tumor.37 When appropriate, surgical removal of the tumor is done without a biopsy, and this provides the necessary tissue for the pathologist.

Some tumors are biopsied during a surgical procedure called an open biopsy.25,70 For those patients, the tumor may be removed at the same time. MRI is generally used to locate the best area to biopsy. For brain tumors located in parts of the brain that are difficult to reach or in areas that are vital for survival, a stereotactic biopsy is preferred. This method uses fine computer-guided instruments and produces less trauma. However, about 2% of stereotactic biopsies result in hemorrhages that impair brain functioning.69 Positron emission tomography (PET), a type of imaging that looks for abnormally functioning cells, is undergoing research to determine whether it can improve biopsy accuracy.

Biomarker-Guided Treatment Decisions

Temozolomide (Temodar), a type of drug called an alkylating agent, causes damage to the DNA of cancer cells. The MGMT gene encodes a DNA repair protein.25 When the MGMT protein is abundant in cancer cells, the cells can repair the damage caused by temozolomide and survive.

In some glioblastomas, the MGMT gene is inactivated in a process called DNA methylation.70 These tumors have very little or no MGMT protein available to repair the damage caused by temozolomide. As a result, these tumors tend to respond well to temozolomide.77

Temozolomide usually has to be given in high doses, and prolonged administration may lead to side effects, which may be more severe in older patients.78-80 Testing a patient’s tumor for MGMT methylation has become a valuable biomarker to predict their response to temozolomide, and can help them and their doctors decide whether they are good candidates for the drug.81-83 Patients without MGMT methylation might be better candidates for other therapies.84-86

Assessing Prognosis

Another part of the diagnostic process involves gathering information on a patient's prognosis, which is an estimation of the likely course of his or her disease. A small group of prognostic factors associated with improved patient outcomes have been identified for patients with glioblastoma87:

  • Age 50 or less88
  • A score of 70 or more on an assessment tool for functional impairment called the Karnofsky Performance Scale (KPS) Index (lower scores indicate greater levels of impairment)88
  • A tumor not located in an “eloquent” brain location, including areas involved in speech, vision, movement, the thalamus, basal ganglia, and internal capsule87,89
  • A tumor that can be completely or almost completely removed in surgery88,90
  • Molecular features of the tumor, such as MGMT methylation or mutations in a gene called IDH170,87,91

6 Participating in a Clinical Trial

Before any new cancer tests or treatments are made available, they must first pass through a series of clinical trials to ensure they are effective and safe in patients. For some patients with glioblastoma, participation in one of these clinical trials may be the best or perhaps only option. Ask your medical team about available clinical trials when they are presenting treatment options and work with them to decide if being part of a clinical trial is right for you.

Clinical trials that eventually lead to approved treatments are conducted in five phases92:

  • Phase 0 trials are preliminary trials that enroll few (10‒15) people to examine how the drug is absorbed, broken down, and excreted in the human body, as predicted from laboratory and animal studies. These trials determine whether further clinical development should proceed.
  • Phase I trials involve a small number of people (around 20‒80). They mostly focus on testing the safety of a drug, and seek to find the highest dose that can be given safely and without risk of adverse effects.
  • If a drug passes phase I, it moves on to a phase II clinical trial. In phase II trials, which involve larger groups of people (100‒300), researchers gather data on how effective the drug is for treating a specific type of disease, and study its safety in more detail.
  • If phase II results are promising, phase III trials are conducted to compare the new drug to standard treatment. These trials usually involve large numbers of people (hundreds or thousands) and are critical for demonstrating the value of the new drug to the Food and Drug Administration (FDA) and the medical community.
  • Lastly, phase IV trials are conducted on already-approved treatments to examine their long-term effects on even larger groups of people. Sometimes phase IV trials examine other potential benefits of the drug or discover additional side effects.

Clinical trials have strict rules on who can participate. For instance, a trial might be restricted to patients who have not yet been treated for their disease or have tumors with a specific characteristic. Each trial lists its rules for participation as “inclusion criteria” and “exclusion criteria,” and these details are included in the clinical trial descriptions found at www.clinicaltrials.gov.

Participation in a trial has some risks, such as unexpected side effects, and the new treatment may not be effective. However, participants may be among the first to have access to cutting-edge treatments and will receive the highest standard of patient care. Regardless of the trial outcome, every participant helps researchers improve treatment options for future patients.

The following websites may be helpful for finding out more about clinical trials and clinical trial participation:

7 Conventional Treatment

Determining the Treatment Approach

Glioblastoma is notoriously difficult to treat effectively, partly because every patient’s tumor has different molecular and cellular characteristics. These characteristics can vary even within the same tumor. Research continues to examine ways to personalize glioblastoma treatment, with the hope of improving outcomes by creating a treatment plan specific for each tumor’s unique characteristics.93-96 Most treatment planning is still based largely on more general characteristics, such as patient age, functional status (KPS Index score), and more recently, MGMT methylation status.35,70,85,97 Initial surgery to remove as much of the tumor as possible is the mainstay of treatment for most people with glioblastoma, followed by radiotherapy and/or chemotherapy.70,98

Surgery and Local Chemotherapy

Surgery is an essential part of glioblastoma treatment.70 Surgical removal (resection) of a glioblastoma tumor can relieve symptoms, extend life, and decrease the need for corticosteroids to reduce brain swelling. The amount of tumor that can be removed through surgery depends on its location, as well as the patient's age and health status. Ideally, surgeons aim for what is referred to as a maximum safe resection, which will remove most or all of the tumor.90,99,100 In some cases, glioblastoma tumor cells spread in different directions so that the tumor may not be a simple solid mass, making total resection impossible.37 Major surgical centers now use a special compound called 5-aminolevulinic acid (5-ALA), colloquially referred to as “pink drink,” which lights up the tumor so the surgeon can see it better. The patient drinks it just before surgery. The surgeon then shines a UV light on the brain tissue during surgery and removes as much of the dyed tissue as possible. This new technique has improved the rate of maximum safe resection.101 When combined with MRI imaging at the time of surgery, both accuracy and precision are greatly improved. The safety profile and documented improvement in outcomes in patients has led experts to advocate for 5-ALA use as standard of care.102

Within three days after surgery, and preferably within the first 24 hours, MRI scans are necessary to determine how much of the tumor was removed. For those unable to undergo MRI, CT scans with and without contrast can be performed.103-105

During surgery, some patients may be treated with a form of chemotherapy delivered locally to the tumor site.70,106 A drug called carmustine is contained in a wafer, and up to eight wafers may be placed into the space where the tumor was. Placing the wafers directly into the brain helps the drug target any remaining tumor cells without damaging healthy cells in other parts of the body.25 The wafers dissolve over time after surgery.107 Although this form of chemotherapy may extend the life of the patient, it can cause complications.108,109 For example, carmustine may interact with some other drugs and increase their toxicity.70 Also, some patients may experience swelling in the brain, seizures, healing problems, or local infections.110,111 Due to its high toxicity and emerging new agents that continue to show promise, the use of implanted carmustine wafers has declined over time.

Systemic Chemotherapy

Unlike carmustine wafers, some other chemotherapeutic drugs are delivered to the whole body through the bloodstream. This is called “systemic” therapy and is accomplished by using pills taken orally or liquids injected into a vein.70 Some patients will only receive one drug, usually the alkylating agent temozolomide. Temozolomide damages the DNA.112 Cancer cells are growing and dividing rapidly and are more sensitive to DNA damage.113,114 Tumors with MGMT methylation may be more sensitive to alkylating agents.37,81 For some patients, several drugs are used to fight the cancer in multiple ways.70

Because systemic chemotherapy can be hard on a patient’s body, the treatments are normally spaced out in a series of cycles, typically two to four weeks each.70 Side effects of systemic chemotherapy depend on the drug, dose, and individual patient. Because chemotherapy drugs typically target rapidly dividing cells, healthy cells that also divide rapidly, such as those in the digestive tract, blood, and hair follicles, may also be affected by the drug. Common side effects of systemic chemotherapy include low blood cell count, loss of appetite, nausea, fatigue, vomiting, diarrhea, hair loss, and mouth sores.115-117

More general information about chemotherapy, including strategies to reduce chemotherapy side effects, is available in the Chemotherapy protocol.

Temozolomide Resistance

Temozolomide has improved glioblastoma treatment outcomes over the past two decades. However, not all cancers are sensitive to temozolomide, and even those that start out sensitive typically develop ways to overcome the drug.118

Finding ways to overcome or prevent temozolomide resistance is a critical area of research. Many of the compounds discussed in the Integrative Interventions section, such as curcumin, quercetin, and resveratrol, are being tested in laboratory studies to see if they can make glioblastoma cells more sensitive to temozolomide.

Repurposed drugs like metformin and cimetidine are also being analyzed in the same way. In one recent study, researchers selected 21 drugs promising or already known to work against various kinds of cancer.119 They tested whether the drugs would make glioblastoma cells more sensitive to temozolomide. One drug called hydroxyurea, a treatment for sickle cell disease and some types of cancer, was clearly the standout. Hydroxyurea sensitized all types of glioblastoma cells and tumors in mice to temozolomide, regardless of the MGMT methylation status.

Another drug called trans sodium crocetinate (TSC) is designed to make tumors more sensitive to temozolomide and radiation, possibly by increasing their oxygen content.120 In a phase II clinical trial, 36% of patients treated with TSC in addition to temozolomide and radiation were still alive after two years, compared to historical survival rates of up to 30% at two years with standard of care.121 Preparation for a phase III clinical trial is currently underway to compare TSC-treated patients to a control group.122

Anti-angiogenesis Therapy

Tumors rely on the growth of new blood vessels, or angiogenesis, to provide nutrition to each cell. A monoclonal antibody called bevacizumab (Avastin) prevents angiogenesis by blocking the vascular endothelial growth factor (VEGF) signaling pathway.70 VEGF signaling is often increased in glioblastoma tumors and contributes to tumor growth. Bevacizumab was approved in 2009 for treatment of recurrent glioblastoma.37 A review study found that bevacizumab could improve patients’ median survival by four months for recurrent glioblastoma, as compared to those not taking the drug.123 Bevacizumab can also lead to reduction in the need for steroids for symptom control.124 Side effects can occur with bevacizumab, and patients must be monitored for excessive bleeding or blood clots.37,123,124

Another anti-angiogenic drug sometimes used when glioblastoma recurs is regorafenib (Stivarga).70 This small molecule inhibits several pathways of survival and growth in glioblastoma cells, including some of those involved in promoting angiogenesis. In addition to regorafenib, many other drugs that target angiogenesis are being researched to determine the best means of thwarting new blood vessel growth in glioblastoma.125

Radiation Therapy

Some glioblastoma tumors cannot be surgically removed. Patients in these situations are treated with radiation therapy.70,126 Additionally, radiation therapy can be used to treat patients after surgery, with the goal of killing any cancer cells that may have been left behind.25 This form of therapy uses high-energy, highly focused rays to damage and destroy cancer cells. Modern radiation therapy uses techniques designed to minimize damage to nearby healthy tissues.127,128 Most patients with glioblastoma receiving radiotherapy will be treated with a method called external beam radiation therapy (EBRT), in which radiation from a large machine passes through the skin and bone and into the brain tissue.70 The treatment machinery is adjusted to deliver radiation as carefully as possible to the tumor area, limiting exposure to healthy tissue. The routine addition of three-dimensional (3D) planning tailored for each person before they begin radiation therapy has reduced the damage to normal brain tissue when compared to older protocols.

For more complete information on radiation therapy techniques and side effects, see the Radiation Therapy protocol.

Tumor-Treating Fields

Tumor-treating fields, or alternating electric field therapy, is a technique that utilizes low-intensity electromagnetic energy to stop cells from dividing.81,129 The treatment uses patches taped to the patient's head and a portable battery-powered device.70,130 The patches must be worn at least 18 hours per day. This technique is relatively safe, and mild-to-moderate local skin irritation is the most commonly reported side effect.81,131

In a 2017 randomized controlled trial, 695 people with glioblastoma were treated with tumor-treating fields along with temozolomide or temozolomide alone. The median time to survival without disease progression was 6.7 months in the tumor-treating field group, as compared with four months in the temozolomide-only group.132 The FDA initially approved tumor-treating fields for patients with recurrent glioblastoma in 2011, and expanded that approval in 2015 to include patients newly diagnosed with glioblastoma.81 Emerging data have shown that some of the benefit from tumor-treating fields may be due to immune stimulation, making its use alongside immunotherapies intriguing.133

Follow-Up and Continuing Care

MRI scans should be conducted 2‒6 weeks after the end of radiation therapy. Additional scans should be performed every 2‒4 months to check for any new brain tumors as early as possible. Those who cannot undergo MRI (such as those with certain types of pacemakers or defibrillators) can receive CT scans with and without contrast. MRI scan interpretation can be challenging due to a phenomenon known as pseudoprogression. As the name implies, what appears to be progression on the MRI image is not actual growth of the tumor. Instead, the area of the tumor appears larger due to the treatment stimulation to that brain region. This can happen in the first 1‒3 months of treatment.70 Whether there is true progression or pseudoprogression is determined by the neuro-oncologist.

Unfortunately, most glioblastomas grow back.37,98 MR spectroscopy, MR perfusion, or PET scan may help confirm recurrences.70 Treatment options for recurrences are similar to options for newly diagnosed disease. Surgery with or without carmustine wafers may be an option for recurrent tumors that are not widespread. In some cases, the goal of surgery is to alleviate symptoms.37 For recurrences, systemic chemotherapy, radiation therapy, bevacizumab, and tumor-treating field therapy may be used.70,81

Supportive Care

Supportive (or palliative) care is not intended to treat the cancer, but may enhance the patient’s quality of life and alleviate symptoms.37,70 Examples of supportive interventions include the use of glucocorticoids, a type of steroid, to reduce swelling in the brain. Additionally, supportive care may involve treating a patient's depression or fatigue, decreasing delirium or agitation, improving cognition, and controlling seizures.134,135 Supportive care may be the only option for patients with advanced or recurrent glioblastoma.

8 Novel and Emerging Strategies

There are several intriguing glioblastoma therapies supported by emerging evidence. One way to access some of these therapies may be through participation in a clinical trial. Ask your medical team about available clinical trial options. Alternatively, innovative physicians familiar with the latest research may be willing to incorporate some of the more widely available off-label drugs described here into a conventional treatment plan.

Immunotherapy

Immunotherapy is a major research focus in the field of oncology. Under healthy conditions, the immune system is able to keep cancer in check.136 However, some cancer cells develop the ability to escape the immune system.137 Once cancer cells that are not vulnerable to immune destruction have established themselves in a person’s body, a tumor can start to form.138

Immunotherapy aims to manipulate the patient’s immune system to enable it to once again attack and eliminate cancer cells.139 Recent advances in using immunotherapy for various cancers opened interest towards applying this strategy for glioblastoma, and a better understanding of the tumor microenvironment is critical for these developments.140 Researchers are investigating many different types of immunotherapies for glioblastoma.141,142

For example, T-cell therapies are being explored. In a phase I study on a 50-year-old man with recurrent glioblastoma143 the investigators created a chimeric antigen receptor–engineered (CAR) T cell. This CAR-T cell was designed to target interleukin-13 receptor alpha 2, a marker expressed by many glioblastoma cells.144 The patient received treatment with these CAR-T cells, which were injected into his brain over a period of 220 days.143 The patient had a dramatic clinical response without any serious negative effects. His original tumors disappeared, and the response continued for 7.5 months. However, his cancer recurred at new sites after treatment was stopped. Given these intriguing findings, the researchers are expanding an ongoing study to administer the CAR-T cells to more patients and are optimizing the treatment.145 CAR-T cells have been designed to target other glioblastoma markers, including HER2 and a form of the epidermal growth factor receptor.146,147 While T-cell therapies have been approved for other cancers, in the case of glioblastoma it appears they may need to be combined with other treatments to give rise to a robust immune response against glioblastoma.8

Checkpoint inhibitors, a type of immunotherapy that helps the immune system recognize cancer cells, can be very effective for several other cancers, but treatment with checkpoint inhibitors after glioblastoma surgery has been disappointing.148 However, studies suggest the timing of immune treatments may influence their success or failure. In one study, a checkpoint inhibitor called nivolumab (Opdivo) was administered before surgery for glioblastoma. While it did not change outcomes for patients undergoing a second surgery, two of the three participants that were newly diagnosed were still alive after 33 and 28 months (at the time of the paper’s publication).149 In another study, T cells that target CMV, the virus that has been associated with glioblastoma, were generated from the patients’ own cells. Of the 25 patients in the study, those who were treated before their first progression fared much better than those who had already progressed at least once since their initial surgery. Several of them had no evidence of progression at 24 months.150

In 2013, research conducted at Duke University Medical Center reported complete clinical responses in patients with recurrent glioblastoma that were treated with a modified poliovirus that had been altered using genetic material from a rhinovirus (a type of virus that causes common colds). This poliovirus-rhinovirus hybrid, called PVSRIPO, is engineered to not infect non-cancerous cells.14,151,152 However, because glioblastoma cells commonly have high levels of a receptor that binds to poliovirus, the poliovirus hybrid can infect them and trigger cell death.153 In addition, molecules released from the dying cells activate immune and inflammatory responses that help destroy the cancer cells.151,154-156 Due to positive reports from early research and lack of effective therapies for glioblastoma, PVSRIPO received “breakthrough therapy” designation from the FDA in May 2016, allowing further investigations to proceed rapidly.153,154,157

A phase I clinical trial designed to identify optimal doses of PVSRIPO for future clinical trials enrolled 61 patients with recurrent high-grade glioblastoma. Varying doses were applied directly to their tumors using catheters. A survival rate of 21% was noted at both 24 and 36 months among those treated with PVSRIPO versus 14% at 24 months and 4% at 36 months in a comparison group of 104 similar patients not treated with this therapy. The non-infectiousness of PVSRIPO was confirmed, and while adverse effects occurred frequently, only one treatment-limiting side effect was observed in a participant receiving the maximum tested dose (the patient developed a grade 4 intracranial hemorrhage).158 Five of the patients from this original phase I study underwent retreatment with more doses of PVSRIPO. Three of the five were alive at 81, 80, and 52 months after the first dose of PVSRIPO; overall, 21% of participants were still alive at 36 months.27

Cancer vaccines, a type of immunotherapy earning much attention in recent years, may be effective against brain tumors.1,159,160 One anti-cancer vaccine in clinical trials, called SurVaxM, targets a protein called survivin.161 Survivin is commonly expressed in glioblastoma cells and normally protects cancer cells from death.162-164 The vaccine is designed to trigger an immune response against survivin, just like a flu vaccine makes the body recognize the virus that causes flu. Early studies showed that the vaccine produced an anti-tumor immune response against gliomas in mice. In a phase I trial of patients with recurrent glioblastoma, SurVaxM was safe and improved outcomes for trial participants.165 The patients on average went 17.6 weeks without worsening of their disease and survived an average of 86.6 weeks.161 The FDA has granted SurVaxM orphan drug status, and a larger trial of SurVaxM in combination with standard therapy is planned for newly diagnosed glioblastoma.166 SurVaxM is also undergoing clinical study for treatment of recurrence in glioblastoma.166 This study is using SurVaxM in combination with an approved immunotherapeutic agent (pembrolizumab), which helps the immune system better recognize cancer cells.167

Rindopepimut (CDX-110) is similar to SurVaxM in that it is a vaccine that targets a particular aberration in glioblastoma. This aberration is in a gene called epidermal growth factor receptor variant III (EGFRvIII), and it occurs in about 60‒70% of glioblastomas. Early studies were promising, but this vaccine did not lead to better outcomes when used alone.168,169 However, studies of rindopepimut combined with maintenance temozolomide or bevacizumab have demonstrated longer times to progression and better overall survival times.170 Vaccines that leverage several targets at once are also undergoing clinical trials, with mixed results.171 

Photodynamic Therapy

Photodynamic therapy uses light, often ultraviolet light, at a specific wavelength to kill tumor cells. This light combines with a chemical “sensitizer” that concentrates in cancer cells but not normal cells. In the case of glioblastoma, 5-ALA, the same dye agent used for lighting up the tumor before surgical removal, is used as the sensitizer. This technique reaches all hard-to-reach areas that extend out from a glioblastoma tumor. It also includes some of the cells that are most resistant to chemotherapy or radiation, such as stem-like glioma cells and dormant cells.172,173 Ideally, the surgeon physically removes as much tumor as possible, then shines the UV light on the brain. Surgery followed by photodynamic therapy has resulted in better outcomes than surgery alone.174 Research is also underway to determine if there are ways to improve 5-ALA uptake into glioblastoma cells using various drug combinations.175 Photodynamic therapy is generally only available in the research setting as of late 2020.174

Targeted Drugs

Targeted drugs bind to a specific site or compound in cancer cells. Binding to the site or “target” interferes with growth signaling pathways. Several genetic abnormalities can occur in glioblastoma, and this provides the “target” that is not found in normal brain cells nearby. Increasingly, these genetic abnormalities are being tested as part of the initial pathology after biopsy or tumor resection. There is one targeted agent currently recommended for recurrent glioblastoma, regorafenib.70 Regorafenib is a small molecule tyrosine kinase inhibitor that targets several cell-signaling pathways involved in cancer pathobiology, including VEGFR1-3, RET, c-kit, PDGFRα and β, among others.176

Many targeted drugs are still undergoing research, in both preclinical and clinical studies.27 There are many potential drugs that may be appropriate in a limited number of patients with glioblastoma, and many of them target specific molecular abnormalities in a person’s glioblastoma.171 Whether a targeted agent is appropriate to consider should be discussed with your neuro-oncologist.

Metformin

Metformin, a first-line drug for diabetes, can pass from the bloodstream into the brain.177 A number of preclinical studies have shown that metformin may inhibit the division and migration of glioblastoma cells.178-183 In laboratory studies, metformin stopped glioblastoma stem cells from dividing,183 and metformin and arsenic trioxide helped differentiate glioblastoma stem cells into non-tumorigenic cells.182

The anti-cancer effects of metformin may result in part from activation of the enzyme AMP-activated protein kinase (AMPK) and inactivation of the transcription factor STAT3.181,184-186 AMPK is an important regulator of glucose and fatty acid metabolism that promotes healthy aging and extends lifespan187,188 while STAT3 controls cell growth and survival and is activated in many cancer types.189,190 Metformin has also been shown to lower levels of a protein, EZHIP, that causes epigenetic changes that contribute to a type of brain tumor called group A posterior fossa ependymoma (PFA), which occurs more often in children than adults.488,489 In one study, metformin suppressed metabolic activity in PFA cells and increased survival of mice bearing xenografts of these deadly brain tumors.488

Metformin may synergize with some existing cancer treatments. For example, in one study, metformin improved the ability of temozolomide to destroy human brain cancer cells.191 A separate study used a type of glioblastoma cells that were not responding to temozolomide. Treatment with metformin made the cells sensitive to temozolomide.192 In mice with experimentally induced glioblastoma, metformin improved the effects of temozolomide, and in cell culture studies, it improved the effects of radiation therapy.193,194 In another study, mice with glioblastoma treated with high-dose metformin combined with temozolomide lived significantly longer than those treated with metformin or temozolomide alone.195 An angiogenesis inhibitor called sorafenib (Nexavar) was also more effective when combined with metformin in laboratory research.179 In another laboratory study, metformin sensitized glioblastoma cells to radiation or radiation combined with temozolomide.196 Additional findings from animal research showed metformin decreased brain swelling and reduced the leakiness of the blood vessels.197

Initial data in human glioblastoma patients have also been encouraging. One study analyzed data from 276 glioblastoma patients treated with either radiation or radiation plus temozolomide. Forty of the patients had diabetes, and 20 of these were taking metformin. Survival time without evidence of disease worsening was significantly longer in diabetics receiving metformin (10 months) than in other diabetics (less than 5 months) and nondiabetics (7 months).198 As of mid-2021, there are five clinical trials (three phase II and two phase I) registered with ClinicalTrials.gov that address the potential benefits of metformin in combination with other therapies in people with glioblastoma.200 Results of these trials will help establish the value of metformin as a component of adjuvant therapy for glioblastoma.

Metformin has also been studied as part of an off-label drug “cocktail” for cancer patients, particularly glioblastoma patients, at the Care Oncology Clinic in London, England.201 The clinic’s combination includes metformin, atorvastatin, mebendazole, and doxycycline, each of which has shown some efficacy against glioblastoma and glioblastoma cell lines in preclinical studies as well as in a rodent model and phase I clinical study.202-206 In the Care Oncology Clinic study, an interim analysis of this retrospective, open-label, single-arm trial found that the addition of the drug combination to standard of care appeared to extend survival in patients with glioblastoma compared with historical standard of care alone. The cohort of patients who received the drug cocktail had a median survival of 27.1 months and 64% of participants survived past two years.201 This compared favorably to historical survival of 14.8‒15.8 months with standard of care.98,487 However, this ongoing study has several key methodological limitations, including lack of direct comparison with a control group as well as biased patient selection. Therefore, much more rigorous study is needed to determine whether this drug combination offers additional survival benefits over standard of care.

Valganciclovir

Valganciclovir is an FDA-approved drug used to treat CMV infection.207 In a phase I/II clinical trial of valganciclovir involving 42 patients with glioblastoma, an exploratory analysis of 22 patients receiving at least six months of antiviral therapy found that 50% were still alive after two years compared with 20.6% of the control group not receiving valganciclovir. After four years, about 27% of patients who received valganciclovir for greater than six months and almost 6% of control participants were still alive.11 In a similar study, researchers compared data from glioblastoma patients treated with valganciclovir and a control group. Both groups received standard conventional therapy and had similar disease characteristics. After two years, 62% of the valganciclovir group and 18% of the control group were still alive. Among the 40 patients who received valganciclovir for at least six months, 70% were still alive after two years.63 In a follow-up study, 102 newly diagnosed glioblastoma patients were given valganciclovir in addition to standard treatment. At two years, 49.8% were still alive compared with 17.3% of control patients at the same center.12 Separately, in a smaller trial of eight glioblastoma patients who had progressing tumors, the addition of valganciclovir resulted in a median survival of 19.1 months versus 12.7 months in those who did not receive it.13

Based on these results, patients with glioblastoma and evidence of CMV-positive tumor tissue should consider consulting with their oncologist to see if they are eligible to receive the treatment protocol described in the aforementioned studies.11,63 The treatment protocol consisted of 900 mg valganciclovir twice daily for three weeks and then 450 mg twice a day. The dose can be adjusted if any side effects arise such as kidney impairment or bone marrow suppression.

Dichloroacetate

Dichloroacetate is an investigational drug that has shown benefits for certain genetic diseases.208 In recent years, dichloroacetate has gained attention for its ability to kill cancer cells and enhance the activity of other cancer therapies.209

Early research has been promising: an open-label phase I trial on 15 adults with grade III or IV gliomas or brain metastases from other cancers found that dichloroacetate treatment was feasible and well-tolerated.210 A similar trial in 24 patients with advanced solid tumors used 28-day cycles of dichloroacetate at different doses and found only mild side effects; this trial was not designed to assess how well the treatment worked.211 This research built on an earlier, smaller trial on five glioblastoma patients treated with dichloroacetate for up to 15 months.212 The authors found evidence of glioblastoma cell death and reduced formation of new blood vessels (angiogenesis) in these patients’ tumors. Studies on cancer cells in the lab have also shown that dichloroacetate increases cancer cell death and decreases angiogenesis, which is necessary for tumors to spread.210,212 Dichloroacetate also has been found to make the inside of the glioblastoma cells dramatically more acidic, which may inhibit their growth.213 Ongoing research into the therapeutic potential of dichloroacetate in solid cancers is likely to focus, at least in part, on finding the best dose, as individual responses vary widely.214,215

Antidepressants

Antidepressant drugs are also being examined for possible effects on glioblastoma cells. For example, fluoxetine (Prozac), a common antidepressant drug, has been shown to selectively kill glioblastoma cells in laboratory experiments.216 Additionally, fluoxetine may reduce the amount of MGMT in glioblastoma cells and make them more sensitive to temozolomide.217 Other antidepressant drugs, such as imipramine (Tofranil) and amitriptyline (Elavil), have been shown to stop glioblastoma stem cells from producing more stem cells.218

Rapamycin and mTOR Inhibition

Rapamycin is an immunomodulating drug first identified in soil samples from Easter Island in the 1970s. Since its discovery, much has been learned about how rapamycin functions in the body. The drug inhibits signaling through a pathway called the mammalian target of rapamycin (mTOR). The mTOR signaling pathway integrates growth signals with cellular metabolism and is involved in many cellular processes, including growth, cell division, protein synthesis, and cell death.219-221 To perform its cellular activities, mTOR functions as part of two distinct multi-protein complexes, mTORC1 and mTORC2, which have different functions and respond differently to rapamycin.221-223 Studies in recent years have identified many interesting properties of the mTOR pathway, and revealed its potential as a target for cancer therapy.

In glioblastoma, increased mTOR signaling has been linked to stem cell proliferation, relapses, and resistance to treatment. In a study that used glioblastoma cells obtained from patients, rapamycin inhibited cell growth, and in mice that had human-derived glioblastomas, it almost doubled the survival time of the animals.222 In another study, rapamycin reduced the proliferation of glioblastoma cancer stem cells and their tumorigenic potential.224

Results of clinical studies using rapamycin have been modest or uncertain.225,226

Rapamycin showed benefits in a phase I clinical trial in certain patients with glioblastoma227 but results from phase II clinical trials were not promising. At least in part, this is explained by the interaction with other signaling pathways.228 Additionally, even though targeting mTOR is a promising strategy for glioblastoma, neither of the two complexes is completely inhibited by rapamycin or rapamycin analogs. However, an experimental compound that inhibited both mTORC1 and mTORC2 together was able to block the growth and migration of glioblastoma cells, underscoring the promise of this approach.223 The combined inhibition of the two complexes was also underscored as a promising therapy by other studies on glioblastoma.229-231

As of the time of this writing, researchers are exploring ways to manipulate the mTOR pathway that might improve outcomes for people with glioblastoma. Existing drugs that target mTOR do not appear well suited as glioblastoma therapies for the time being.

GSK-3ß Inhibition

GSK-3, an enzyme responsible for many reactions in cells, is integrally involved in many aspects of tumor pathobiology, including proliferation, invasion, and survival. While targeting this enzyme has had mixed results for various types of tumors, it appears that inhibiting the GSK-3beta (GSK-3β) form specifically has an anti-glioblastoma effect.232

A drug combination called CLOVA (cimetidine, lithium, olanzapine, and valproate) was tested in a small 2017 study on seven glioblastoma patients. The study found that the CLOVA cocktail led to longer-than-expected survival. The mechanism by which this drug cocktail improved survival was thought to involve inhibition of GSK-3β.155

Kenpaullone is drug that specifically inhibits GSK-3β and has shown early promise in the context of glioblastoma. Kenpaullone was discovered through a screening process of over a thousand chemicals in search of one that effectively inhibits GSK-3β.233 By downregulating the activity of GSK-3β, kenpaullone inhibits the proliferation of glioma cells while encouraging their self-destruction pathways (apoptosis). Targeting GSK-3β also results in the reduction of the stem cell like features of glioblastoma, a characteristic tied to its aggressiveness. The effect of kenpaullone may also complement the therapeutic effects of temozolomide through reduction of the cells’ ability to produce MGMT.234

All-trans Retinoic Acid (ATRA)

Carotenoids, which are precursors of vitamin A, and retinoids, which are derivatives structurally similar to vitamin A, have shown anti-oxidative properties and protective effects against certain cancer types.235-238 The anti-cancer effects of one retinoid, called all-trans retinoic acid (ATRA), have been examined in several studies.239-241 ATRA, either alone or in combination with a drug called rapamycin, stimulated glioblastoma cancer stem cells to change into specialized cells and slowed their movement.242 Another study found that ATRA disrupted the movement of stem-like glioma cells and decreased production of chemicals that stimulate blood vessel formation.243 A recent study found that ATRA enhanced the effects of temozolomide on human glioblastoma cells.244 The treatment of human glioblastoma cells with ATRA or another retinoid, called 13-cis retinoic acid or isotretinoin, made the cells more likely to die when exposed to the chemotherapy drug paclitaxel (Taxol).245 Bexarotene (Targretin), a retinoid used to treat lymphoma,246 inhibited the migration of glioblastoma cells and changed the expression of several cancer-related genes towards a more beneficial profile. The compound also killed tumor cells in a mouse model of glioblastoma.247

The beneficial effects of retinoids have been explored in clinical trials that enrolled patients with glioblastoma.248-250 Isotretinoin has been explored in several studies as maintenance therapy, intended to help delay tumor recurrence. One retrospective analysis found patients taking isotretinoin lived an average of approximately 25 months without disease progression compared to an average of approximately eight months in those not taking isotretinoin.251 The most common side effects were skin-related.248

Multi-Drug Combinations (Repurposed Drugs)

Glioblastoma tumors are well known to eventually become resistant to every chemotherapy drug used to treat them. Since singular pathway inhibition is universally overcome by glioblastoma cells, there is interest in finding multiple drugs that may be used in combination to simply dampen the growth rather than eradicate the disease. The use of multiple repurposed drugs to simultaneously inhibit many pathways at once is undergoing investigation. These are usually combinations of less toxic drugs that are approved for other disease processes. One ongoing combination under investigation is called CUSP9, and uses nine drugs (aprepitant, artesunate, auranofin, captopril, celecoxib, disulfiram (DSF), itraconazole, ritonavir, and sertraline) to overcome over a dozen pathways of drug resistance to temozolomide.252,253

One combination that has shown encouraging results in animals is known as FTT (fasudil, tranilast [Rizaben], and temozolomide).254 Fasudil is a vasodilating agent used for stroke victims and tranilast is an anti-allergy drug approved for use in Asian countries that acts on a key inflammatory pathway called TGF-β.254 This combination suppressed tumor growth and increased the time of survival more so than temozolomide alone in animals given glioblastoma. Clinical studies of this combination are needed before any conclusions can be made on its use in humans.

Inhaled Cannabidiol

Cannabidiol (CBD), the second most abundant phytocannabinoid present in cannabis, has garnered interest for its potential benefits in cancer treatment.492,493 CBD can act as both an agonist and antagonist of the endocannabinoid CB1 and CB2 receptors, although it displays low affinity as an agonist.494 CB1 receptors are primarily found in the central nervous system; CB2 receptors are primarily found on cells in the immune system and hematopoietic cells and are thought to elicit the immune-modulatory effects of CBD.492,494 Both CB1 and CB2 receptors have been found to be expressed in many cancer types, and several preclinical studies suggest CBD may have therapeutic efficacy in the context of some cancers.492,495 Since CBD does not produce psychoactive effects, in contrast to Δ9-tetrahydrocannabinol (Δ9-THC), it has remained a focus of intense research.

In a preclinical study, researchers utilized a mouse model of glioblastoma to examine the effect of inhaled CBD (10mg/day) on the tumor microenvironment, which is vital for tumor survival and progression.496-498 They found that inhalation of CBD limited tumor growth in addition to altering the dynamics of the tumor microenvironment by repressing P-selectin, apelin, interleukin-8 (IL-8), and blocking indoleamine 2,3-dioxygenase (IDO), a key protein that allows tumor cells to escape detection by the immune system. In fact, high expression of IDO is significantly associated with poor prognosis of tumors.499 Inhaled CBD also increased CD103 expression, promoted CD8+ T-cell immune responses, and decreased innate lymphoid cells within the tumor.498

9 Dietary and Lifestyle Considerations

The American Cancer Society and American Brain Tumor Association have several dietary and lifestyle recommendations for cancer patients. Good nutrition can help patients manage the side effects of cancer treatment, maintain energy,255 avoid infections,256 and even fight the disease. In general, patients’ diets should be rich in a variety of vegetables and healthy sources of protein and unsaturated fats.255,257,258 Diets high in colorful fruits and vegetables contain high amounts of phytochemicals. Many of these phytochemicals are broken down by bacteria in the gut to form compounds that can cross the blood-brain barrier and influence neuronal function.259 One class of phytochemicals, called polyphenols, may confer some protection against the development of glioma by modulating many of the inflammatory pathways involved in glioma formation and growth.260 For some patients, an exercise program may improve mood and quality of life.261

Ketogenic Diet

The ketogenic diet emphasizes healthy fats and proteins with very little carbohydrates (typically less than 20 grams net carbohydrates daily).262-264 This diet is sometimes recommended to reduce seizure frequency in children and adults with epilepsy, but may also be helpful in those with glioblastomas because these tumors are known to rely on carbohydrates for energy.262,265

A ketogenic diet has been found to control tumor growth and prolong survival in animal studies.265-267 Other studies have found that the diet may boost immune response to tumor cells and provide benefits when used in combination with other treatments, such as radiation.268 In humans, the diet leads to lower circulating glucose levels, which is associated with better outcomes in those with glioblastoma, and it is generally well tolerated and safe.269 Blood tests can be used to check how well the diet is reducing blood glucose levels and increasing ketone levels.270 A review of 24 human studies found 42% of the studies suggested there may be direct anti-tumor effects of the ketogenic diet.271 Quality of life improvements, including sense of overall wellbeing, fewer seizures, and better neurological function was more consistently reported. Several phase I or II interventional trials have been conducted or are underway to investigate whether a ketogenic diet can improve outcomes for people with glioblastoma.272-274

Some research has suggested restricting caloric intake may enhance the effects of a ketogenic diet.275,276 In one small study of patients with glioblastoma, only the few participants that lost at least 10% of their body weight derived any benefit from the ketogenic diet.277 Caloric restriction may be accomplished by reducing daily intake or by intermittent fasting. However, patients with advanced cancer should work with a nutritional oncologist to ensure they are consuming adequate nutrition.275

Ketogenic diets may have the dual effect of protecting normal cells and sensitizing cancer cells to therapeutic radiation.278 At the heart of the diet is the production of ketones, which can be used by cells in lieu of glucose to derive energy. One of the primary ketones produced in the body, beta-hydroxybutyrate, functions as a histone deacetylase (HDAC) inhibitor. Aberrant HDAC signaling occurs in several types of cancer, including gliomas. Some researchers have advocated for more widespread use of this therapeutic diet in glioblastoma.279,280

Case Report: Ketogenic Diet, Integrative Interventions, and Repurposed Drugs in a Glioblastoma Patient

A case report published in 2018 combined many of the emerging treatments and natural agents mentioned throughout this protocol into a single protocol designed to affect metabolism of the tumor.281 First, the patient, a 38-year-old male, fasted for 72 hours before surgery. Unfortunately, there was residual tumor on imaging after the surgery. The patient went on to receive standard of care treatments along with several prescriptive and integrative therapies designed to control the growth of glioblastoma through inducing cellular stress and withholding preferred energy sources. The interventions included a vitamin and mineral blend, vitamin D (5,000 IU/day), methylfolate (1,000 mg/day), epigallocatechin gallate (EGCG) (400 mg/day), metformin (1,000 mg/day), levetiracetam (1,500 mg/d), and chloroquine phosphate (150 mg/day). The patients also underwent hyperbaric oxygen therapy (60 minutes/session, 5 days weekly, at 2.5 times normal atmospheric pressure). After a 21-day course of a low-calorie ketogenic diet, the patient went on to increase his calories somewhat but maintain a reduced-calorie ketogenic diet for nine months. The ketogenic diet was done under the watchful eye of his medical care team. At 24 months, his team published their results. The patient was healthy with excellent quality of life. He lost about 19% of his baseline body weight and his cancer remained in remission.

Coffee and Tea Consumption

Coffee and tea have also been explored as a potential dietary intervention for reducing the risk of developing gliomas. In a large study of participants from 10 European countries, daily intake of 100 mL (about half a cup) or more of coffee or tea was associated with a lower risk of developing glioma. The association was slightly stronger in men. This same beneficial effect was reported in another study that examined the intake of coffee and tea in people from the United States. This US-based study reported that those drinking five or more cups of coffee and/or tea per day were less likely to develop gliomas than those who drank less than one cup per day.282

Coffee contains many phytochemicals that may have anti-cancer effects.283 One of the most interesting compounds is a polyphenol called chlorogenic acid, which has been shown to inhibit glioblastoma cell growth in laboratory studies.284,285 Other compounds in coffee include kahweol and cafestol, which have been shown in animals to increase the activity of MGMT, which is commonly silenced in glioblastoma cells.286

Similarly, one of the compounds found in tea, called epigallocatechin gallate (EGCG), has reversed the silencing of MGMT in cell culture experiments.287 EGCG has also been shown to improve the efficacy of temozolomide in a mouse glioblastoma model.288,289 In addition, EGCG has been shown to reduce invasion, lessen proliferation, and may enhance other therapies such as carmustine.290 One caveat to green tea is that it may interfere with a class of drugs called tyrosine kinase inhibitors, as suggested by one study using bortezomib for multiple myeloma.291 While some authors have raised valid criticisms of this study,292 until there is solid evidence refuting interference with cancer drugs, high-dose green tea should be taken with caution alongside tyrosine kinase inhibitors and only under the guidance of an oncologist.

10 Nutrients

Melatonin

In humans, the natural hormone melatonin is involved in the sleep-wake cycle and in endocrine function. Disturbances in sleep-wake cycles lead to daytime fatigue, and this disturbance is one of the most common symptoms in people with brain tumors.293 Melatonin can stimulate the immune system and help fight inflammation.294,295 For some patients with insomnia, melatonin can help improve their quality of sleep.296-298 As we gain more insight into melatonin’s molecular mechanisms, it is becoming clear that it affects normal cells and cancer cells differently, resulting in selective harm to cancer cells without harming normal cells.299

Recent laboratory evidence has shown that melatonin may inhibit the viability and self-renewal of glioblastoma stem-like cells.294 In a study on glioblastoma stem-like cells isolated from patient surgical samples, melatonin affected cellular signaling pathways involved in cell survival and division.300 Melatonin may block glioblastoma cells from invading new areas by inhibiting genes involved in tissue invasion and new blood vessel formation.301 Melatonin may also interfere with the aberrant energy production within glioblastoma cells that otherwise allows them to divide rapidly.302

In laboratory studies, melatonin boosted the effects of chemotherapy drugs, including temozolomide, indicating it may be especially helpful for patients undergoing conventional treatment.303 In addition, melatonin may complement the anti-cancer effects of repurposed drugs such as metformin, statins, and anti-inflammatories.304 It has been shown that glioblastoma cells reduce the amount of melatonin produced within their own mitochondria, providing them with a survival advantage.305

In one early clinical trial, 30 patients with glioblastoma were randomized to either radiation therapy plus oral melatonin (20 mg per day) or radiation therapy alone. After one year, six of 14 patients taking melatonin and only one of 16 patients in the control group were still alive. Those taking melatonin had less hair loss and infections as well. The authors also noted that side effects of radiation were less frequent in the melatonin group.306 The potential for melatonin’s use in glioblastoma needs further clinical trials, but its potential for improved outcomes and better quality of life in those with glioblastoma is promising.21

Vitamin D

Vitamin D and some of its metabolites have been shown to stop glioblastoma cells from dividing in a laboratory setting.307-309 In one study, glioblastoma cells that had a mutation in a gene called p53, which is frequently dysregulated in glioblastoma,310 were more vulnerable to death when exposed to vitamin D.311

Intriguingly, one study found that levels of the vitamin D receptor were increased in glioblastoma tissue samples compared with non-cancerous brain tissue, and that this was associated with improved survival.312 Patients with the vitamin D receptor present in their tumors had a better outcome in one retrospective analysis.313 A laboratory study showed that vitamin D enhanced the toxicity of temozolomide against glioblastoma cells. Also, combined treatment with temozolomide and vitamin D prolonged survival and reduced tumor progression in a rat model of glioblastoma.314 Alfacalcidol, an analogue of vitamin D, was used in a clinical study (at a dose of 0.04 mcg/kg) alongside standard treatments for primary brain tumors. There were 11 patients (10 with glioblastoma and one with astrocytoma, grade III). Three of the 11 patients (2 glioblastoma and 1 astrocytoma) showed a very favorable response with continued reduction in tumor and complete clinical remission for 7, 5, and 4 years. The authors proposed that 20% of those with high-grade gliomas may respond favorably to alfacalcidol.315 Among the many mechanisms possible for the effects of vitamin D and its analogues, there is evidence of stopping glioblastoma cells from dividing and inducing senescence. Vitamin D and analogues of the natural form are continuing to undergo extensive research.312

Selenium

Selenium is an essential trace element316 and an integral player in brain health, including brain cancers.317 The first clinical evidence of a link between selenium and brain cancers came when it was found that selenium levels in the blood were significantly lower in patients with brain malignancies than in healthy individuals.318 Clinical studies have not yet confirmed the benefit of selenium supplementation for glioblastoma patients, but laboratory studies suggest selenium may reduce some of the negative effects of chemotherapies while making cancer cells more sensitive to chemotherapies.319 For instance, sodium selenite decreased cell proliferation and caused cell death in several types of human glioblastoma cells.320 In another laboratory study, sodium selenite inhibited the proliferation of human glioblastoma cells and rat glioma cells.321 A mixture of nutrients that contained several ingredients, including selenium, lysine, proline, ascorbic acid, and green tea extract, significantly decreased the ability of glioma cells to invade through a gelatinous material used in the laboratory to study tumor dissemination.322 A study that chemically linked selenium to temozolomide reported that the new compound was effective against temozolomide-resistant glioma cells; also, in human glioblastoma cells, the new compound caused DNA breaks and killed the cells more effectively than temozolomide alone.323

Boswellia

There are naturally-occurring plant compounds under investigation for their anti-tumor properties, such as boswellic acids, found in the gum resin of Boswellia plants, better known as frankincense.324-326 Boswellic acids have shown promise in cell culture experiments and animal studies against several cancer types, including colorectal cancer, glioma, prostate cancer, pancreatic cancer, and leukemia.327 In particular, these potent compounds can induce cell death, suppress inflammation, decrease tissue invasion and blood vessel formation, and inhibit signaling pathways that stimulate cancer development.327,328 In a mouse model of brain tumors, boswellia reduced tumor growth by normalizing several aspects of aberrant metabolism in the glioma tissue.329 In another cell model, boswellic acids stopped the process of division in glioma cells.330

A recent study described experiments designed to determine whether boswellic acids could enhance the anti-cancer effects of standard therapies, such as temozolomide or radiation. The treatment of human glioblastoma cells with boswellic acids led to cell death. When boswellic acids were used in combination with temozolomide, afatinib (Gilotrif), or radiation, a combined effect greater than the sum of their separate effects was observed, indicating boswellic acids could be a promising integrative therapy for patients with glioblastoma.324,331,332

Boswellic acids are also helpful in reducing brain swelling, which may develop as a result of brain tumors or their treatment with radiation therapy.333-335 One study tested the effects of H15, a boswellic acid-containing extract from the gum resin of the Boswellia serrata plant, on brain swelling in 12 patients with brain tumors. Swelling was reduced in two of seven glioblastoma patients.335 In a second study, 44 patients with brain tumors took either 4,200 mg boswellia extract daily or placebo while undergoing radiation therapy. The boswellia extract group had a significant decrease in brain swelling compared with placebo. An over 75% reduction of swelling was seen in 60% of patients receiving the extract versus 26% of patients receiving placebo.336 In an uncontrolled study, a highly absorbable lecithin-based boswellia extract was used in 20 patients with glioblastoma who were receiving standard of care. Each of them consumed 4,500 mg boswellia daily for up to 34 weeks. Swelling around the brain was assessed at 4,12, 22, and 34 weeks after surgery and steroid consumption was tracked. Steroid use was either reduced, stable, or not needed for most of the patients over the course of the study. Two patients achieved dramatic reduction of edema that likely contributed to their improved outcomes. The authors noted that the anti-inflammatory effects of boswellia on the brain likely led to reduction of steroids and their side effects as well as better outcomes.337

Curcumin

Curcumin, derived from the Curcuma longa plant, is a component of the spice turmeric.338 Several laboratory studies have examined the cellular effects of curcumin on glioblastoma cells. Curcumin affects several cancer pathways necessary for cell division, survival, invasion, and metastasis.339-342 Curcumin may reduce or even eliminate glioblastoma stem cells, which are notoriously unaffected by chemotherapy, by reducing their number, killing them, or changing them into a less-dangerous cell type.20,338,343,344

One study used a form of curcumin bound to an antibody to help target curcumin to the glioblastoma cells and nearby microglia, a type of support cell in the central nervous system. The combination was used to treat mice with glioblastoma. Remission of the glioblastoma was noticed in half of the animals. Laboratory analyses indicated curcumin killed the glioblastoma cells and improved the ability of the microglial cells to kill nearby cancer cells.345 In another study in mice, animals were transplanted with human glioblastoma cells and treated with curcumin. Curcumin crossed into the brain, inhibited the formation of new blood vessels and the breakdown of surrounding tissue (extracellular matrix) that otherwise allows for tumor growth.346

In another animal glioblastoma model, rats that were given curcumin and treated with radiation lived longer than when either curcumin or radiation was given as a single agent. The authors concluded that curcumin may be an effective radiosensitizing agent for gliomas.347 However, the dose needed to reach the brain to replicate these rodent studies may be too high for humans to achieve.348 Novel delivery forms of curcumin and curcumin analogues are being researched to overcome this hurdle and appear promising.349,350

There is also evidence that curcumin may enhance the efficacy of some chemotherapy drugs.339 In a laboratory study on glioblastoma cells, curcumin increased the anti-proliferation, anti-migration, and cell death activities of nimustine hydrochloride, a chemotherapy drug widely used for treating glioblastoma. This combined treatment might be a promising therapeutic approach.351 Curcumin may also enhance the effectiveness of temozolomide, and much like combining curcumin with radiation, novel ways to enhance this effect through improved curcumin uptake into the brain are being sought.352

Curcumin may also have an effect on cancer cells through its ability to increase the production of ceramide, a type of fat molecule (lipid) found within the membranes of cells.353-355 This finding is important because increased ceramide has been found to sensitize glioma cells to chemotherapy.356

L-arginine

L-arginine administration has been found, in a controlled clinical trial, to influence metabolic processes and induce radiosensitivity in tumors that have metastasized to the brain. L-arginine acts as a precursor for nitric oxide (NO), which was shown to induce chemo- and radiosensitivity in solid and hypoxic tumors by increasing tumor blood flow and decreasing tumor oxygen consumption. NO is synthesized by NO synthases (NOS), which are found in three isoforms; of these isoforms, nitric oxide synthase 2 (NOS2) has been shown to be transcriptionally upregulated in a cell line of triple-negative breast cancer as well as in patients with brain metastasis due to non-small cell lung carcinoma.490

In a preliminary study of five patients with brain metastases, 10 grams of oral L-arginine in water was capable of increasing plasma L-arginine levels by an average of 42%. L-arginine did not alter tumor blood flow but decreased tumor blood volume and lactate concentration compared with placebo, highlighting the suppressive effect of L-arginine on tumor metabolism.490

To examine the effect of L-arginine on radiation therapy, the researchers performed a clinical study and recruited 63 patients with unresectable brain metastases that were randomized to receive 10 grams oral L-arginine or placebo 1 hour prior to each fraction of radiation therapy with a median follow up of five months (ranging from 1 to 55 months). There was a significantly greater overall response rate, symptomatic response rate, complete neurological response, and proportion of patients free from neurological progression in those treated with L-arginine compared with placebo.490 These novel results demonstrated that oral L-arginine could be an effective strategy to induce radiosensitivity in patients with brain metastases.

L-arginine is also capable of suppressing tumor metabolism and DNA repair in preclinical in vitro and in vivo models; these effects may contribute to its radiosensitizing effects. In triple-negative breast cancer cell lines, L-arginine administration significantly reduced several measures of tumor metabolism.490 Further, L-arginine decreased DNA damage repair in cancer cells in vitro and in mice in vivo without altering the amount of radiation-induced DNA damage in tumor-infiltrating lymphocytes, indicating that L-arginine is unlikely to harm immune cells.490,491 Arginine’s suppression of tumor metabolism was mediated by NOS2 and prevented by pretreatment with a NOS2 inhibitor prior to L-arginine administration.490

Resveratrol

Resveratrol is found in certain plants where its role is to protect the plant from infectious microbes.357-359 Nuts, berries, grapes, red wine, and Japanese knotweed are excellent sources of resveratrol.360 Resveratrol is being explored as a potential anti-cancer treatment because some evidence suggests it helps prevent instability of the genome, reduces the ability of gliomas to invade nearby tissue, and improves the effectiveness of some conventional treatments.361,362

Resveratrol’s potential as a therapeutic agent for brain tumors hinges in part on its ability to cross the blood-brain barrier as well as its ability to inhibit proliferation, migration, and cell survival.357 In one study, resveratrol inhibited the growth of human glioblastoma cells and caused cell death in a dose-dependent manner.363 It also inhibited the growth of glioblastoma stem-like cells and suppressed the growth of glioblastoma in a mouse model.364 In another study, resveratrol was able to prevent the cellular changes needed for glioblastoma cells to become invasive.365 Resveratrol inhibited a signaling pathway in these cells and suppressed the production of a protein involved in cellular invasion.366 In a laboratory study that used several glioblastoma cell types, resveratrol inhibited cellular movement and invasiveness by activating a major signaling pathway.22

Resveratrol may also increase the sensitivity of cancer cells to temozolomide and radiation. In one study, glioblastoma-initiating cells were isolated from two patients with glioblastoma. Resveratrol sensitized these cells to temozolomide.367 In in vitro studies and mouse models, temozolomide more effectively induced cell death and inhibited cell migration when used together with resveratrol.367,368 Resveratrol may overcome temozolomide resistance by reducing the amount of MGMT in the resistant cells.369,370 In a glioma stem cell line resistant to radiation, resveratrol increased the sensitivity of the cells to radiation.371 Resveratrol also increased the ability of the chemotherapy drug paclitaxel to kill glioblastoma cells.372

Quercetin

Quercetin is a naturally occurring plant flavonoid with many potential anti-cancer properties.373,374 Multiple laboratory experiments have demonstrated that quercetin can kill human glioblastoma cells.375 Quercetin may also inhibit the ability of glioblastoma cells to metastasize,376,377 reduce their viability,377,378 decrease their ability to proliferate and migrate,379 and inhibit blood vessel formation.376 Other research found quercetin may increase the sensitivity of glioblastoma cells to temozolomide and radiation.380,381 Quercetin may also enhance the effects of chloroquine, an antimalarial drug with promise as an anti-cancer agent for glioblastoma.382

Green Tea and EGCG

Epigallocatechin-3-gallate (EGCG) is a green tea flavonoid with known anti-cancer, antioxidant, and anti-inflammatory activities.383-385 In laboratory studies that used human glioblastoma cell lines, exposure to EGCG contributed to cell death.383,386 EGCG targets several cellular events mediated by matrix metalloproteinases, including some pathways that control cellular migration.387 EGCG can also inhibit a protein that makes glioblastoma cells more resistant to chemotherapy and blocks their death.388 In human glioblastoma stem-like cells, EGCG synergized the effects of temozolomide.288 A study found that this synergy may be due to reversal of the resistance mechanism, namely MGMT, within the glioblastoma cells.389

EGCG and other catechins from green tea may fight cancer partly through their ability to inhibit the activity of an important cellular signaling pathway.390 In two different human glioblastoma cell types, EGCG activated cell death pathways. Interestingly, EGCG did not have this effect on healthy human brain cells.391 One study showed that green tea led to senescence of glioblastoma cells without harming normal cells.392 Another cell study suggested that the dose derived from drinking green tea may prevent brain tumor development, but glioblastoma cell death is only achieved from much higher concentrations of green tea.393 Research in mice with glioblastoma is also encouraging. EGCG significantly improved the therapeutic effects of temozolomide, and the combination extended survival of the mice compared with temozolomide alone.289 In a separate study in mice implanted with human gliomas, EGCG slowed tumor growth by interfering with the aberrant metabolic pathway of gliomas.394 Interestingly, application of EGCG to colon cancer cells harboring a mutation in IDH1 led to decreased proliferation.395 Whether this will hold true for IDH1-mutated gliomas has not been studied as of the time of this writing.

Chrysin

Chrysin, a naturally occurring flavonoid found in honey, propolis, and many plants, may fight inflammation and other processes involved in cancer development.396 Chrysin promoted cell death in studies of several glioblastoma cell lines.397,398 Another study found chrysin reduced the mitochondrial function of glioblastoma cells and decreased the production of a protein involved in tumor invasion.399 In a mouse xenograft model, chrysin inhibited the proliferation, migration, and invasion of glioblastoma cells and suppressed tumor growth.400 In an in vitro study, an extract of propolis inhibited the growth of human glioblastoma cells in a dose- and time-dependent manner and enhanced the effects of temozolomide.401

Apigenin

Another plant-derived compound called apigenin inhibited cellular pathways involved in glioblastoma cell proliferation and survival. Apigenin treatment caused the cells to stop at a certain point in their cell division process.402 Apigenin also powerfully suppressed the invasiveness of glioblastoma stem-like cells.403 This is a significant finding because stem-like cells can self-renew and are resistant to radiotherapy and chemotherapy.404,405 In human glioma cells, apigenin reduced the production of TGF-β1, a signaling molecule involved in migration, invasion, and the formation of blood vessels.406 Importantly, apigenin may not have the same effects on normal cells. One study found apigenin activated cell death pathways in two different human glioblastoma cell lines, but not in normal human astrocytes.391 Central to any cancer growth is the ability to evade immune destruction. Apigenin was able to reduce glioma cell migration and restore immune attraction toward glioma cells, a necessary component of their eventual destruction.407 Apigenin has numerous anti-cancer actions across a diverse array of tumors.408

Phytoestrogens

Phytoestrogens are plant compounds that are similar in structure to the hormone estrogen.409 Soy beans, flaxseed, and nuts are all good sources.410,411 Despite their similarities, phytoestrogens appear to play a different role in glioblastoma than the estrogens naturally produced by the body. This may be because they act differently inside cells. Endogenous estrogens have an unpredictable effect in glioblastomas, leading either to growth promotion or inhibition, depending on numerous factors.408,412,413 In contrast, plant-derived phytoestrogens have been shown to exert beneficial effects in glioblastoma.414

In a mouse model of human glioblastoma, a phytoestrogen called genistein inhibited tumor growth after 10 days of treatment. Cellular and molecular analyses suggested genistein slowed tumor growth by decreasing the formation of new blood vessels in the tumor.415 Another study found genistein may decrease the proliferation of glioblastoma cells by stopping their division and lowering the activity of telomerase, an enzyme that cancer cells need to protect the ends of their chromosomes and survive.416,417 In a preclinical experiment, genistein inhibited a signaling pathway used by glioblastoma cells even at very low concentrations.418 Glioblastoma cells that were exposed to genistein had more sensitivity to the killing capacity of therapeutic radiation doses, lowering the dose needed to kill the cells.419 In another experiment, genistein lessened the invasive potential of glioblastoma cells.370

Daidzein is another phytoestrogen. One study found that daidzein can help activate cellular pathways involved in cell death in glioblastoma cells. Healthy brain cells were not affected by this treatment.420 A lesser-known phytoestrogen called biochanin A (found in red clover421 and other plants) enhanced the anti-cancer effects of temozolomide.422

Honokiol

Honokiol is a natural bioactive polyphenol extracted from the bark of the tree Magnolia officinalis. Honokiol has demonstrated anti-inflammatory, anti-microbial, and anti-cancer effects in laboratory studies.423,424 Researchers have reported that honokiol can inhibit the division of glioblastoma cells and cancer stem-like cells422,425 and kill glioblastoma cells by several mechanisms.426,427 The ability of honokiol to cause glioblastoma cell death may result, at least in part, from its ability to stimulate a protein that causes cell death and inhibit a protein that prevents cell death.428 Another study found honokiol inhibited the interaction between human glioblastoma cells and cells that line the blood vessels, suggesting it may inhibit the spread of tumor cells via the bloodstream.429 In glioblastoma cell culture experiments, honokiol and a similar compound called magnolol were more effective at killing cancer cells when used together.423

Honokiol is of particular interest for treatment of glioblastoma because studies in mice suggest the compound can cross the blood-brain barrier.430 In a mouse model of human glioblastoma, honokiol caused cell death and significantly prolonged survival of the mice.424 A number of genes involved in regulating the cell cycle were activated in the treated mice. In a similar study, the combination of honokiol and magnolol inhibited tumor progression and killed cancer cells more efficiently than the chemotherapy drug temozolomide.423

In an experiment using honokiol and a disulfiram/copper complex in a liposomal delivery system, tumor regression via immune attack was observed in a mouse model.431 When combined with temozolomide, several studies have shown honokiol may complement the drug’s ability to kill glioblastoma cells.432-434

Polyunsaturated Fatty Acids

Several types of polyunsaturated fatty acids (PUFAs) have been studied for the treatment of glioblastoma.435 Treating glioblastoma cells with docosahexaenoic acid (DHA), an omega-3 PUFA, led to several cellular and molecular changes that indicate cell death. The authors followed up with an additional experiment in mice with glioblastomas. The mice were altered to express an enzyme that converts omega-6 PUFAs to omega-3 PUFAs. The increase in omega-3 PUFAs was associated with a decrease in tumor volume.436 When various types of glioma cells were exposed to different PUFAs, including arachidonic acid, gamma linolenic acid (GLA), and DHA, the expression of certain genes involved in cell death increased.437 Open-label clinical studies have suggested GLA may be effective against malignant gliomas.438,439 In patients with glioma, delivering GLA directly into the tumor was found to be safe, and in some cases, led to tumor regression. Several participants survived without new symptoms for up to two years.440

Milk Thistle

Silibinin (silybin) is a biologically active compound in extracts from the seeds of the herb milk thistle (Silybum marianum ).441,442 Silibinin is capable of affecting many of the classic characteristics or “hallmarks” of cancer that allow the cells to grow and spread unchecked.443 Studies show silibinin caused glioblastoma cells to enter into self-destructive mode through at least two separate mechanisms.444,445 In another study, silibinin inhibited the invasive features of highly invasive glioblastoma cells.446 Another strategy tested silibinin in combination with luteolin, another plant-derived compound. The combination inhibited the growth of glioblastoma cells more effectively than temozolomide, slowed cell migration, and caused glioblastoma cells and glioblastoma stem cells to die.447,448

Using several different glioma cell lines, silibinin increased the toxicity of temozolomide on the cells, improving the drug’s cell killing ability.449 Silibinin also worked well in combination with arsenic trioxide, a drug approved for treatment of a form of leukemia.450,451 In glioblastoma cells, the combination of silibinin and arsenic trioxide slowed tumor cell metabolism and increased cell death.452 A study found that silibinin increased the accumulation of arsenic inside glioblastoma cells treated with arsenic trioxide. When silibinin was combined with chrysin, arsenic trioxide levels built up inside glioblastoma cells, while the cells were simultaneously hampered in their defenses against it.453

Vitamin E

The term “vitamin E” refers to eight compounds in nature; four tocotrienols and four tocopherols. The tocotrienols, alpha-, beta-, gamma-, and delta-, may help fight cancer and inflammation.454,455 They target many molecular paths used by cancers, including survival mechanisms, new blood vessel formation, proliferation and invasion.456 In a laboratory study, alpha-, gamma-, and delta-tocotrienols inhibited the growth of human glioblastoma cells and caused DNA breaks. Delta-tocotrienol killed the cells more effectively than alpha- and gamma-tocotrienol.457 Delta-tocotrienol also worked well in combination with extracts from the Tabernaemontana corymbosa plant, a traditional cancer treatment in Bangladesh,458 and extracts from plants in the genus Ficus.455 In vitro, tocopherols showed anti-glioma effects by controlling cell cycle progression in glioma cells. In this study, gamma-tocopherol was found to exhibit “the most potent and specific control” over glioma cells’ progression through the cell cycle.459 Similarly, another in vitro study also found gamma-tocopherol was more potent than alpha-tocopherol in inhibiting proliferation, adhesion, and migration in human glioma cells.460

Ellagic Acid

Ellagic acid, a natural compound found in many fruits and plants, may also have health benefits for glioblastoma patients. In general, ellagic acid is being studied for its ability to lessen spread of cancers to distant organs and reduce the production of new blood vessels.461 In human glioblastoma cells, ellagic acid inhibited the viability and proliferation of the cells and damaged their DNA. The authors then confirmed these results in mice with glioblastoma and found that ellagic acid inhibited signaling pathways involved in cancer cell proliferation and invasion.462 Another study reported ellagic acid dramatically reduced levels of proteins that protect tumor cells from death.463 A root extract of Leonurus sibiricus L., a traditional medicinal plant found in China, Japan, Korea, Vietnam, and southern Siberia, contains ellagic acid and several other polyphenolic compounds. The extract effectively killed human glioblastoma cells by regulating genes involved in cell death.464 When glioblastoma cells were treated with ellagic acid combined with temozolomide, they exhibited a reduction in the invasive and angiogenic capabilities of the cancer cells.465 Ellagic acid may also complement bevacizumab, an approved anti-angiogenesis drug for glioblastoma. In a cell study, ellagic acid combined with bevacizumab led to reduced viability of cells.466 In the same study, ellagic acid led to downregulation of MGMT expression, which increases glioma cells’ sensitivity to the cytotoxic effects of temozolomide.

Chlorogenic Acid

Chlorogenic acid, a phenolic compound found in coffee, green tea, apples, and pears, has promising anti-cancer potential for many cancers, including brain tumors.467 The compound inhibited the growth of glioblastoma cells and reduced the growth of glioblastomas in mice. Some of the immune cells in the tumors of these treated mice were changed to a form that can more readily destroy tumor cells.284 Another study found chlorogenic acid inhibited cell migration and the secretion of a protein implicated in tumor invasion.285 In a double-blind placebo-controlled trial, chlorogenic acid improved cognitive function in healthy participants.468 This is intriguing given the cognitive difficulties that are common during and after treatment for glioblastoma.

Ginseng

Ginseng (Panax ginseng) root contains compounds called ginsenosides. One ginsenoside in particular, R3, has been shown to have an additive anti-tumor effect when combined with temozolomide in an animal model of glioblastoma.469 In another study using several glioma cell lines that were resistant to temozolomide through production of MGMT, R3 was able to block MGMT production and reinstate sensitivity of the cells to temozolomide.470 This study also showed ginsenoside R3 prevented the changes in glioma cells that allow them to invade nearby tissue. It appears that at least some R3’s anti-glioblastoma properties are via its antioxidant properties and induction of senescence in glioma cells.471 R3 ginsenoside is being studied as an anti-cancer agent due to its well defined role in blocking new blood vessel development in many types of cancer, including gliomas.472

Olive Leaf

Olive leaf (Olea europaea) extract exhibited anti-proliferative effects in glioma cells.473 Experiments on glioblastoma cell lines suggest olive leaf extract can prevent the differentiation ability of glioblastoma stem cells.474 The anti-glioblastoma effects of olive leaf extract are seen when used alone in high dose, and it has an additive effect in stopping glioma cell growth alongside temozolomide.475 Whole leaf extract has generally been studied, and single compounds within the olive leaf, such as oleuropein, also show promise476; however, most of the research is preliminary as of late 2020.

Black Cumin

Black cumin (Nigella sativa) seed has been used in traditional herbal medicine for over 2,000 years.477 While whole seed extracts have shown effective medicinal qualities, the compound thymoquinone in black cumin has garnered the most interest as an anti-cancer compound.478,479

Thymoquinone has been shown to affect dozens of pathways inside cancer cells, including those involved in several hallmarks of cancer: immortality, invasion, metastasis, apoptosis, and cellular division.480 The anti-glioblastoma effects include increasing cell death and creating cellular stress within glioblastoma cells.481,482 The benefits of thymoquinone may also extend to preventing ongoing brain-related symptoms as data suggest neuroprotective and anti-seizure effects.483

Niclosamide (Niclocide) is another anti-glioblastoma compound of interest from black cumin.484 This compound is thought to be responsible for the traditional use of black cumin seeds in treating parasites and worms in the gut. Niclosamide is also an approved anti-parasitic agent worldwide due to its reliability in killing human worms such as tapeworm.485 In one experiment, the combination of thymoquinone and niclosamide was able to lessen the invasive properties of a glioblastoma cell model. The combination also had a similar effect in a rodent model that used implanted human glioblastoma cells.486

2022

  • Jan: Added section on inhaled cannabidiol (CBD) to Novel & Emerging Strategies

2021

  • Nov: Added section on L-arginine to Nutrients
  • Oct: Updated section on metformin in Novel & Emerging Strategies
  • Jan: Updated section on metformin in Novel & Emerging Strategies
  • Jan: Comprehensive update & review

Disclaimer and Safety Information

This information (and any accompanying material) is not intended to replace the attention or advice of a physician or other qualified health care professional. Anyone who wishes to embark on any dietary, drug, exercise, or other lifestyle change intended to prevent or treat a specific disease or condition should first consult with and seek clearance from a physician or other qualified health care professional. Pregnant women in particular should seek the advice of a physician before using any protocol listed on this website. The protocols described on this website are for adults only, unless otherwise specified. Product labels may contain important safety information and the most recent product information provided by the product manufacturers should be carefully reviewed prior to use to verify the dose, administration, and contraindications. National, state, and local laws may vary regarding the use and application of many of the therapies discussed. The reader assumes the risk of any injuries. The authors and publishers, their affiliates and assigns are not liable for any injury and/or damage to persons arising from this protocol and expressly disclaim responsibility for any adverse effects resulting from the use of the information contained herein.

The protocols raise many issues that are subject to change as new data emerge. None of our suggested protocol regimens can guarantee health benefits. Life Extension has not performed independent verification of the data contained in the referenced materials, and expressly disclaims responsibility for any error in the literature.

  1. Paolillo M, Boselli C, Schinelli S. Glioblastoma under Siege: An Overview of Current Therapeutic Strategies. Brain sciences. Jan 16 2018;8(1):15-15. doi:10.3390/brainsci8010015
  2. Alexander BM, Cloughesy TF. Adult Glioblastoma. J Clin Oncol. Jul 20 2017;35(21):2402-2409. doi:10.1200/JCO.2017.73.0119
  3. Arevalo AST, Erices JI, Uribe DA, et al. Current Therapeutic Alternatives and New Perspectives in Glioblastoma Multiforme. Curr Med Chem. 2017;24(25):2781-2795. doi:10.2174/0929867324666170303122241
  4. Bianco J, Bastiancich C, Jankovski A, des Rieux A, Preat V, Danhier F. On glioblastoma and the search for a cure: where do we stand? Cellular and molecular life sciences : CMLS. Jul 2017;74(13):2451-2466. doi:10.1007/s00018-017-2483-3
  5. Polivka J, Jr., Polivka J, Holubec L, et al. Advances in Experimental Targeted Therapy and Immunotherapy for Patients with Glioblastoma Multiforme. Anticancer research. Jan 2017;37(1):21-33. doi:10.21873/anticanres.11285
  6. Anjum K, Shagufta BI, Abbas SQ, et al. Current status and future therapeutic perspectives of glioblastoma multiforme (GBM) therapy: A review. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie. Aug 2017;92:681-689. doi:10.1016/j.biopha.2017.05.125
  7. Razmara A. Commentary: 3 Senators, 3 Votes-Glioblastoma's Uncanny Historical Parallels. Neurosurgery. Feb 1 2018;82(2):E55-E57. doi:10.1093/neuros/nyx545
  8. Stylli SS. Novel Treatment Strategies for Glioblastoma. Cancers. Oct 8 2020;12(10):1-11. doi:10.3390/cancers12102883
  9. Dziurzynski K, Chang SM, Heimberger AB, et al. Consensus on the role of human cytomegalovirus in glioblastoma. Neuro Oncol. Mar 2012;14(3):246-55. doi:10.1093/neuonc/nor227
  10. Barami K. Oncomodulatory mechanisms of human cytomegalovirus in gliomas. Journal of clinical neuroscience : official journal of the Neurosurgical Society of Australasia. Jul 2010;17(7):819-23. doi:10.1016/j.jocn.2009.10.040
  11. Stragliotto G, Rahbar A, Solberg NW, et al. Effects of valganciclovir as an add-on therapy in patients with cytomegalovirus-positive glioblastoma: a randomized, double-blind, hypothesis-generating study. International journal of cancer Journal international du cancer. Sep 1 2013;133(5):1204-13. doi:10.1002/ijc.28111
  12. Stragliotto G, Pantalone MR, Rahbar A, Bartek J, Soderberg-Naucler C. Valganciclovir as Add-on to Standard Therapy in Glioblastoma Patients. Clin Cancer Res. Aug 1 2020;26(15):4031-4039. doi:10.1158/1078-0432.CCR-20-0369
  13. Stragliotto G, Pantalone MR, Rahbar A, Soderberg-Naucler C. Valganciclovir as Add-On to Standard Therapy in Secondary Glioblastoma. Microorganisms. Sep 24 2020;8(10):1-11. doi:10.3390/microorganisms8101471
  14. Brown MC, Dobrikova EY, Dobrikov MI, et al. Oncolytic polio virotherapy of cancer. Cancer. Nov 1 2014;120(21):3277-86. doi:10.1002/cncr.28862
  15. Inman S. Cure. FDA Accelerates Development of Polio Virus Treatment for Brain Cancer. Updated 5/17/2016. Accessed 6/9/2017, http://www.curetoday.com/articles/fda-accelerates-development-of-polio-virus-treatment-for-brain-cancer
  16. Abbruzzese C, Matteoni S, Signore M, et al. Drug repurposing for the treatment of glioblastoma multiforme. Journal of Experimental and Clinical Cancer Research. 2017;36(1):169-169. doi:10.1186/s13046-017-0642-x
  17. Kast RE, Karpel-Massler G, Halatsch ME. Can the therapeutic effects of temozolomide be potentiated by stimulating AMP-activated protein kinase with olanzepine and metformin? Br J Pharmacol. Nov 2011;164(5):1393-6. doi:10.1111/j.1476-5381.2011.01320.x
  18. Berg J. A Paradigm Shift in Glioblastoma Treatment and Research: A Multi-mechanistic, Multi-agent Approach to Target Glioblastoma Multiforme. Journal of Advanced Medical Sciences and Applied Technologies. 2017;2(4):323-323. doi:10.18869/nrip.jamsat.2.4.323
  19. Park MN, Song HS, Kim M, et al. Review of Natural Product-Derived Compounds as Potent Antiglioblastoma Drugs. Biomed Res Int. 2017;2017:8139848. doi:10.1155/2017/8139848
  20. Ryskalin L, Biagioni F, Busceti CL, Lazzeri G, Frati A, Fornai F. The Multi-Faceted Effect of Curcumin in Glioblastoma from Rescuing Cell Clearance to Autophagy-Independent Effects. Molecules (Basel, Switzerland). Oct 20 2020;25(20):1-17. doi:10.3390/molecules25204839
  21. Moretti E, Favero G, Rodella LF, Rezzani R. Melatonin's Antineoplastic Potential Against Glioblastoma. Cells. Mar 3 2020;9(3):599-599. doi:10.3390/cells9030599
  22. Xiong W, Yin A, Mao X, Zhang W, Huang H, Zhang X. Resveratrol suppresses human glioblastoma cell migration and invasion via activation of RhoA/ROCK signaling pathway. Oncology letters. Jan 2016;11(1):484-490. doi:10.3892/ol.2015.3888
  23. Association AABT. Brain Tumor Education / Brain Tumor Statistics. American Brain Tumor Association; 2014.
  24. Association AABT. Diet and nutrition during treatment. American Brain Tumor Association; 2014.
  25. Institute NCINC. Adult Central Nervous System Tumors Treatment (PDQ®): Health Professional Version. National Cancer Institute; 2018. p. 1-51.
  26. Lee JH, Lee JE, Kahng JY, et al. Human glioblastoma arises from subventricular zone cells with low-level driver mutations. Nature. Aug 2018;560(7717):243-247. doi:10.1038/s41586-018-0389-3
  27. Wen PY, Weller M, Lee EQ, et al. Glioblastoma in adults: a Society for Neuro-Oncology (SNO) and European Society of Neuro-Oncology (EANO) consensus review on current management and future directions. Neuro Oncol. Aug 17 2020;22(8):1073-1113. doi:10.1093/neuonc/noaa106
  28. Costa RB, Costa R, Kaplan J, et al. A Rare Case of Glioblastoma Multiforme with Osseous Metastases. Case reports in oncological medicine. 2017;2017:2938319. doi:10.1155/2017/2938319
  29. Wu W, Zhong D, Zhao Z, Wang W, Li J, Zhang W. Postoperative extracranial metastasis from glioblastoma: a case report and review of the literature. World journal of surgical oncology. Dec 29 2017;15(1):231. doi:10.1186/s12957-017-1300-7
  30. Seo YJ, Cho WH, Kang DW, Cha SH. Extraneural metastasis of glioblastoma multiforme presenting as an unusual neck mass. Journal of Korean Neurosurgical Society. Mar 2012;51(3):147-50. doi:10.3340/jkns.2012.51.3.147
  31. Ostrom QT, Bauchet L, Davis FG, et al. The epidemiology of glioma in adults: a "state of the science" review. Neuro Oncol. Jul 2014;16(7):896-913. doi:10.1093/neuonc/nou087
  32. Ostrom QT, Gittleman H, Truitt G, Boscia A, Kruchko C, Barnholtz-Sloan JS. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2011-2015. Neuro Oncol. Oct 1 2018;20(suppl_4):iv1-iv86. doi:10.1093/neuonc/noy131
  33. Thakkar JP, Dolecek TA, Horbinski C, et al. Epidemiologic and molecular prognostic review of glioblastoma. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. Oct 2014;23(10):1985-96. doi:10.1158/1055-9965.EPI-14-0275
  34. NBTS. National Brain Tumor Society. Glioblastoma Facts & Figures. Accessed 12/7/2020, https://braintumor.org/take-action/about-gbm/#:~:text=More%20than%2013%2C000%20Americans%20are,succumb%20to%20glioblastoma%20every%20year
  35. Weller M, Stupp R, Hegi M, Wick W. Individualized targeted therapy for glioblastoma: fact or fiction? Cancer journal (Sudbury, Mass). Jan-Feb 2012;18(1):40-4. doi:10.1097/PPO.0b013e318243f6c9
  36. Bielecka J, Markiewicz-żukowska R. The influence of nutritional and lifestyle factors on glioma incidence. MDPI AG; 2020. p. 1-20.
  37. Davis ME. Glioblastoma: Overview of Disease and Treatment. Clinical journal of oncology nursing. Oct 1 2016;20(5 Suppl):S2-8. doi:10.1188/16.CJON.S1.2-8
  38. Urbanska K, Sokolowska J, Szmidt M, Sysa P. Glioblastoma multiforme - an overview. Contemporary oncology (Poznan, Poland). 2014;18(5):307-12. doi:10.5114/wo.2014.40559
  39. Felini MJ, Olshan AF, Schroeder JC, et al. Reproductive factors and hormone use and risk of adult gliomas. Cancer Causes Control. Feb 2009;20(1):87-96. doi:10.1007/s10552-008-9220-z
  40. Andersen L, Friis S, Hallas J, Ravn P, Kristensen BW, Gaist D. Hormonal contraceptive use and risk of glioma among younger women: a nationwide case-control study. Br J Clin Pharmacol. Apr 2015;79(4):677-84. doi:10.1111/bcp.12535
  41. Lan YL, Zou S, Wang X, et al. Update on the therapeutic significance of estrogen receptor beta in malignant gliomas. Oncotarget. Oct 6 2017;8(46):81686-81696. doi:10.18632/oncotarget.20970
  42. Ostrom QT, Gittleman H, Farah P, et al. CBTRUS statistical report: Primary brain and central nervous system tumors diagnosed in the United States in 2006-2010. Neuro Oncol. Nov 2013;15 Suppl 2:ii1-56. doi:10.1093/neuonc/not151
  43. Rice T, Lachance DH, Molinaro AM, et al. Understanding inherited genetic risk of adult glioma - a review. Neuro-oncology practice. Mar 2016;3(1):10-16. doi:10.1093/nop/npv026
  44. Bondy ML, Scheurer ME, Malmer B, et al. Brain tumor epidemiology: consensus from the Brain Tumor Epidemiology Consortium. Cancer. Oct 1 2008;113(7 Suppl):1953-68. doi:10.1002/cncr.23741
  45. Hanif F, Muzaffar K, Perveen K, Malhi SM, Simjee Sh U. Glioblastoma Multiforme: A Review of its Epidemiology and Pathogenesis through Clinical Presentation and Treatment. Asian Pacific journal of cancer prevention : APJCP. Jan 1 2017;18(1):3-9. doi:10.22034/APJCP.2017.18.1.3
  46. McNeill KA. Epidemiology of Brain Tumors. Neurologic clinics. Nov 2016;34(4):981-998. doi:10.1016/j.ncl.2016.06.014
  47. Davis F, Il'yasova D, Rankin K, McCarthy B, Bigner DD. Medical diagnostic radiation exposures and risk of gliomas. Radiation research. Jun 2011;175(6):790-6. doi:10.1667/RR2186.1
  48. Wiedmann MKH, Brunborg C, Di Ieva A, et al. The impact of body mass index and height on the risk for glioblastoma and other glioma subgroups: a large prospective cohort study. Neuro Oncol. Jul 1 2017;19(7):976-985. doi:10.1093/neuonc/now272
  49. Kitahara CM, Wang SS, Melin BS, et al. Association between adult height, genetic susceptibility and risk of glioma. Int J Epidemiol. Aug 2012;41(4):1075-85. doi:10.1093/ije/dys114
  50. Little RB, Nabors LB, Olson JJ, et al. Older age at the completion of linear growth is associated with an increased risk of adult glioma. Cancer Causes Control. Jul 2017;28(7):709-716. doi:10.1007/s10552-017-0871-5
  51. Kabat GC, Rohan TE. Adiposity at different periods of life and risk of adult glioma in a cohort of postmenopausal women. Cancer epidemiology. 2018;54:71-74.
  52. Sergentanis TN, Tsivgoulis G, Perlepe C, et al. Obesity and Risk for Brain/CNS Tumors, Gliomas and Meningiomas: A Meta-Analysis. PLoS One. 2015;10(9):e0136974. doi:10.1371/journal.pone.0136974
  53. Dai Z-F, Huang Q-L, Liu H-P. Different body mass index grade on the risk of developing glioma: a meta-analysis. Chinese Neurosurgical Journal. 2015;1(1):1-6.
  54. Carlberg M, Hardell L. Decreased survival of glioma patients with astrocytoma grade IV (glioblastoma multiforme) associated with long-term use of mobile and cordless phones. International journal of environmental research and public health. Oct 16 2014;11(10):10790-805. doi:10.3390/ijerph111010790
  55. Xu F, Bai Q, Zhou K, et al. Age-dependent acute interference with stem and progenitor cell proliferation in the hippocampus after exposure to 1800 MHz electromagnetic radiation. Electromagnetic biology and medicine. 2017;36(2):158-166. doi:10.1080/15368378.2016.1233886
  56. Kaplan S, Deniz OG, Onger ME, et al. Electromagnetic field and brain development. Journal of chemical neuroanatomy. Sep 2016;75(Pt B):52-61. doi:10.1016/j.jchemneu.2015.11.005
  57. Baan R, Grosse Y, Lauby-Secretan B, et al. Carcinogenicity of radiofrequency electromagnetic fields. The Lancet Oncology. Jul 2011;12(7):624-6. doi:10.1016/s1470-2045(11)70147-4
  58. Non-ionizing radiation, Part 2: Radiofrequency electromagnetic fields. IARC Monogr Eval Carcinog Risks Hum. 2013;102(Pt 2):1-460.
  59. Yang M, Guo W, Yang C, et al. Mobile phone use and glioma risk: A systematic review and meta-analysis. PLoS One. 2017;12(5):e0175136. doi:10.1371/journal.pone.0175136
  60. Havas M. When theory and observation collide: Can non-ionizing radiation cause cancer? Environ Pollut. Feb 2017;221:501-505. doi:10.1016/j.envpol.2016.10.018
  61. Carlberg M, Hardell L. Evaluation of Mobile Phone and Cordless Phone Use and Glioma Risk Using the Bradford Hill Viewpoints from 1965 on Association or Causation. Biomed Res Int. 2017;2017:9218486. doi:10.1155/2017/9218486
  62. CDC. Centers for Disease Control and Prevention. Cytomegalovirus (CMV) and Congenital CMV Infection. Updated 8/18/2020. Accessed 12/7/2020, https://www.cdc.gov/cmv/index.html
  63. Soderberg-Naucler C, Rahbar A, Stragliotto G. Survival in patients with glioblastoma receiving valganciclovir. The New England journal of medicine. Sep 5 2013;369(10):985-6. doi:10.1056/NEJMc1302145
  64. Rahbar A, Orrego A, Peredo I, et al. Human cytomegalovirus infection levels in glioblastoma multiforme are of prognostic value for survival. Journal of clinical virology : the official publication of the Pan American Society for Clinical Virology. May 2013;57(1):36-42. doi:10.1016/j.jcv.2012.12.018
  65. Garcia-Martinez A, Alenda C, Irles E, et al. Lack of cytomegalovirus detection in human glioma. Virol J. Nov 7 2017;14(1):216. doi:10.1186/s12985-017-0885-3
  66. McFaline-Figueroa JR, Wen PY. The Viral Connection to Glioblastoma. Current infectious disease reports. Feb 2017;19(2):5. doi:10.1007/s11908-017-0563-z
  67. Batash R, Asna N, Schaffer P, Francis N, Schaffer M. Glioblastoma Multiforme, Diagnosis and Treatment; Recent Literature Review. Curr Med Chem. 2017;24(27):3002-3009. doi:10.2174/0929867324666170516123206
  68. Reni M, Mazza E, Zanon S, Gatta G, Vecht CJ. Central nervous system gliomas. Critical reviews in oncology/hematology. May 2017;113:213-234. doi:10.1016/j.critrevonc.2017.03.021
  69. DeAngelis LM. Tumors of the central nervous system and intracranial hypertension and Hypotension. In: Cecil RL, Goldman L, Schafer AI, eds. 24th ed. ed. Elsevier/Saunders/; 2012:1246-1256.
  70. NCCN. National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology. Central Nervous System Cancers. Updated 7/25/2016. Accessed 12/8/2020, https://www.nccn.org/professionals/physician_gls/pdf/cns.pdf
  71. Rees JH. Diagnosis and treatment in neuro-oncology: an oncological perspective. The British journal of radiology. Dec 2011;84 Spec No 2:S82-9. doi:10.1259/bjr/18061999
  72. Mullen KM, Huang RY. An Update on the Approach to the Imaging of Brain Tumors. Curr Neurol Neurosci Rep. Jul 2017;17(7):53. doi:10.1007/s11910-017-0760-z
  73. Abrigo JM, Fountain DM, Provenzale JM, et al. Magnetic resonance perfusion for differentiating low-grade from high-grade gliomas at first presentation. The Cochrane database of systematic reviews. Jan 22 2018;1(1):CD011551. doi:10.1002/14651858.CD011551.pub2
  74. Kamson DO, Mittal S, Buth A, et al. Differentiation of glioblastomas from metastatic brain tumors by tryptophan uptake and kinetic analysis: a positron emission tomographic study with magnetic resonance imaging comparison. Molecular imaging. Jul-Aug 2013;12(5):327-37.
  75. Fink KR, Fink JR. Imaging of brain metastases. Surg Neurol Int. 2013;4(Suppl 4):S209-19. doi:10.4103/2152-7806.111298
  76. Neska-Matuszewska M, Bladowska J, Sasiadek M, Zimny A. Differentiation of glioblastoma multiforme, metastases and primary central nervous system lymphomas using multiparametric perfusion and diffusion MR imaging of a tumor core and a peritumoral zone-Searching for a practical approach. PLoS One. 2018;13(1):e0191341. doi:10.1371/journal.pone.0191341
  77. Thomas A, Tanaka M, Trepel J, Reinhold WC, Rajapakse VN, Pommier Y. Temozolomide in the Era of Precision Medicine. Cancer Res. Feb 15 2017;77(4):823-826. doi:10.1158/0008-5472.CAN-16-2983
  78. Lee CY. Strategies of temozolomide in future glioblastoma treatment. OncoTargets and therapy. 2017;10:265-270. doi:10.2147/OTT.S120662
  79. Saito K, Mukasa A, Narita Y, et al. Toxicity and outcome of radiotherapy with concomitant and adjuvant temozolomide in elderly patients with glioblastoma: a retrospective study. Neurologia medico-chirurgica. 2014;54(4):272-9. doi:10.2176/nmc.oa2012-0441
  80. Straube C, Scherb H, Gempt J, et al. Does age really matter? Radiotherapy in elderly patients with glioblastoma, the Munich experience. Radiation oncology (London, England). Apr 28 2017;12(1):77. doi:10.1186/s13014-017-0809-9
  81. Fernandes C, Costa A, Osorio L, et al. Current Standards of Care in Glioblastoma Therapy. In: De Vleeschouwer S, ed. Glioblastoma. Codon Publications; 2017:197-241.
  82. Snyder J, Walbert T. Managing Glioblastoma in the Elderly Patient: New Opportunities. Oncology (Williston Park, NY). Jun 15 2017;31(6):476-83.
  83. Seystahl K, Wick W, Weller M. Therapeutic options in recurrent glioblastoma--An update. Critical reviews in oncology/hematology. Mar 2016;99:389-408. doi:10.1016/j.critrevonc.2016.01.018
  84. Malmström A, Grønberg BH, Marosi C, et al. Temozolomide versus standard 6-week radiotherapy versus hypofractionated radiotherapy in patients older than 60 years with glioblastoma: the Nordic randomised, phase 3 trial. The Lancet Oncology. 2012;13(9):916-926. doi:10.1016/s1470-2045(12)70265-6
  85. Thon N, Kreth S, Kreth FW. Personalized treatment strategies in glioblastoma: MGMT promoter methylation status. OncoTargets and therapy. Sep 27 2013;6:1363-72. doi:10.2147/OTT.S50208
  86. Hau E, Shen H, Clark C, Graham PH, Koh ES, K LM. The evolving roles and controversies of radiotherapy in the treatment of glioblastoma. Journal of medical radiation sciences. Jun 2016;63(2):114-23. doi:10.1002/jmrs.149
  87. Theeler BJ, Gilbert MR. Advances in the treatment of newly diagnosed glioblastoma. BMC Med. Dec 8 2015;13(1):293. doi:10.1186/s12916-015-0536-8
  88. Lacroix M, Abi-Said D, Fourney DR, et al. A multivariate analysis of 416 patients with glioblastoma multiforme: prognosis, extent of resection, and survival. Journal of neurosurgery. Aug 2001;95(2):190-8. doi:10.3171/jns.2001.95.2.0190
  89. Awad AW, Karsy M, Sanai N, et al. Impact of removed tumor volume and location on patient outcome in glioblastoma. Journal of neuro-oncology. Oct 2017;135(1):161-171. doi:10.1007/s11060-017-2562-1
  90. Li YM, Suki D, Hess K, Sawaya R. The influence of maximum safe resection of glioblastoma on survival in 1229 patients: Can we do better than gross-total resection? Journal of neurosurgery. Apr 2016;124(4):977-88. doi:10.3171/2015.5.JNS142087
  91. Chen C, Shi Y, Li Y, et al. A glycolysis-based ten-gene signature correlates with the clinical outcome, molecular subtype and IDH1 mutation in glioblastoma. Journal of genetics and genomics = Yi chuan xue bao. Nov 20 2017;44(11):519-530. doi:10.1016/j.jgg.2017.05.007
  92. Kummar S, Rubinstein L, Kinders R, et al. Phase 0 clinical trials: conceptions and misconceptions. Cancer journal (Sudbury, Mass). May-Jun 2008;14(3):133-7. doi:10.1097/PPO.0b013e318172d6f3
  93. Matteoni S, Abbruzzese C, Villani V, et al. The influence of patient sex on clinical approaches to malignant glioma. Elsevier Ireland Ltd; 2020. p. 41-47.
  94. Bryukhovetskiy I, Pak O, Khotimchenko Y, Bryukhovetskiy A, Sharma A, Sharma HS. Personalized therapy and stem cell transplantation for pro-inflammatory modulation of cancer stem cells microenvironment in glioblastoma: Review. Academic Press Inc.; 2020:67-98.
  95. Loong HH, Wong AM, Chan DT, et al. Patient-derived tumor organoid predicts drugs response in glioblastoma: A step forward in personalized cancer therapy? Journal of clinical neuroscience : official journal of the Neurosurgical Society of Australasia. Aug 2020;78:400-402. doi:10.1016/j.jocn.2020.04.107
  96. Rucco M, Viticchi G, Falsetti L. Towards Personalized Diagnosis of Glioblastoma in Fluid-Attenuated Inversion Recovery (FLAIR) by Topological Interpretable Machine Learning. Mathematics. 2020;8(5):770-770. doi:10.3390/math8050770
  97. Colombo MC, Giverso C, Faggiano E, Boffano C, Acerbi F, Ciarletta P. Towards the Personalized Treatment of Glioblastoma: Integrating Patient-Specific Clinical Data in a Continuous Mechanical Model. PLoS One. 2015;10(7):e0132887. doi:10.1371/journal.pone.0132887
  98. Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. The New England journal of medicine. Mar 10 2005;352(10):987-96. doi:10.1056/NEJMoa043330
  99. Roder C, Bisdas S, Ebner FH, et al. Maximizing the extent of resection and survival benefit of patients in glioblastoma surgery: high-field iMRI versus conventional and 5-ALA-assisted surgery. European journal of surgical oncology : the journal of the European Society of Surgical Oncology and the British Association of Surgical Oncology. Mar 2014;40(3):297-304. doi:10.1016/j.ejso.2013.11.022
  100. Kuhnt D, Becker A, Ganslandt O, Bauer M, Buchfelder M, Nimsky C. Correlation of the extent of tumor volume resection and patient survival in surgery of glioblastoma multiforme with high-field intraoperative MRI guidance. Neuro Oncol. Dec 2011;13(12):1339-48. doi:10.1093/neuonc/nor133
  101. Hadjipanayis CG, Widhalm G, Stummer W. What is the surgical benefit of utilizing 5-aminolevulinic acid for fluorescence-guided surgery of malignant gliomas? : Lippincott Williams and Wilkins; 2015. p. 663-673.
  102. Chohan MO, Berger MS. 5-Aminolevulinic acid fluorescence guided surgery for recurrent high-grade gliomas. Journal of neuro-oncology. Feb 2019;141(3):517-522. doi:10.1007/s11060-018-2956-8
  103. Pirzkall A, McGue C, Saraswathy S, et al. Tumor regrowth between surgery and initiation of adjuvant therapy in patients with newly diagnosed glioblastoma. Neuro Oncol. Dec 2009;11(6):842-52. doi:10.1215/15228517-2009-005
  104. Sanghvi D. Post-treatment imaging of high-grade gliomas. The Indian journal of radiology & imaging. Apr-Jun 2015;25(2):102-8. doi:10.4103/0971-3026.155829
  105. Kumar N, Kumar P, Angurana SL, et al. Evaluation of outcome and prognostic factors in patients of glioblastoma multiforme: A single institution experience. Journal of neurosciences in rural practice. Aug 2013;4(Suppl 1):S46-55. doi:10.4103/0976-3147.116455
  106. Zhang YD, Dai RY, Chen Z, Zhang YH, He XZ, Zhou J. Efficacy and safety of carmustine wafers in the treatment of glioblastoma multiforme: a systematic review. Turk Neurosurg. 2014;24(5):639-45. doi:10.5137/1019-5149.JTN.8878-13.1
  107. Mangraviti A, Tyler B, Brem H. Interstitial chemotherapy for malignant glioma: Future prospects in the era of multimodal therapy. Surg Neurol Int. 2015;6(Suppl 1):S78-84. doi:10.4103/2152-7806.151345
  108. Lara-Velazquez M, Al-Kharboosh R, Jeanneret S, et al. Advances in Brain Tumor Surgery for Glioblastoma in Adults. Brain sciences. Dec 20 2017;7(12)doi:10.3390/brainsci7120166
  109. Bock HC, Puchner MJ, Lohmann F, et al. First-line treatment of malignant glioma with carmustine implants followed by concomitant radiochemotherapy: a multicenter experience. Neurosurg Rev. Oct 2010;33(4):441-9. doi:10.1007/s10143-010-0280-7
  110. Sabel M, Giese A. Safety profile of carmustine wafers in malignant glioma: a review of controlled trials and a decade of clinical experience. Current medical research and opinion. Nov 2008;24(11):3239-57. doi:10.1185/03007990802508180
  111. Giese A, Bock HC, Kantelhardt SR, Rohde V. Risk management in the treatment of malignant gliomas with BCNU wafer implants. Central European neurosurgery. Nov 2010;71(4):199-206. doi:10.1055/s-0029-1242775
  112. Erasimus H, Gobin M, Niclou S, Van Dyck E. DNA repair mechanisms and their clinical impact in glioblastoma. Mutation research Reviews in mutation research. Jul-Sep 2016;769:19-35. doi:10.1016/j.mrrev.2016.05.005
  113. O'Connor MJ. Targeting the DNA Damage Response in Cancer. Mol Cell. Nov 19 2015;60(4):547-60. doi:10.1016/j.molcel.2015.10.040
  114. Baskar R, Dai J, Wenlong N, Yeo R, Yeoh KW. Biological response of cancer cells to radiation treatment. Frontiers in molecular biosciences. 2014;1:24. doi:10.3389/fmolb.2014.00024
  115. Hershman D, Calhoun E, Zapert K, Wade S, Malin J, Barron R. Patients' Perceptions of Physician-Patient Discussions and Adverse Events with Cancer Therapy. Archives of drug information. Sep 2008;1(2):70-78. doi:10.1111/j.1753-5174.2008.00011.x
  116. Stein A, Voigt W, Jordan K. Chemotherapy-induced diarrhea: pathophysiology, frequency and guideline-based management. Therapeutic advances in medical oncology. Jan 2010;2(1):51-63. doi:10.1177/1758834009355164
  117. Mustian KM, Devine K, Ryan JL, et al. Treatment of Nausea and Vomiting During Chemotherapy. US oncology & hematology. 2011;7(2):91-97. doi:10.17925/ohr.2011.07.2.91
  118. Ramirez YP, Weatherbee JL, Wheelhouse RT, Ross AH. Glioblastoma multiforme therapy and mechanisms of resistance. Pharmaceuticals (Basel). Nov 25 2013;6(12):1475-506. doi:10.3390/ph6121475
  119. Teng J, Hejazi S, Hiddingh L, et al. Recycling drug screen repurposes hydroxyurea as a sensitizer of glioblastomas to temozolomide targeting de novo DNA synthesis, irrespective of molecular subtype. Neuro Oncol. Apr 9 2018;20(5):642-654. doi:10.1093/neuonc/nox198
  120. Sheehan J, Cifarelli CP, Dassoulas K, Olson C, Rainey J, Han S. Trans-sodium crocetinate enhancing survival and glioma response on magnetic resonance imaging to radiation and temozolomide. Journal of neurosurgery. Aug 2010;113(2):234-9. doi:10.3171/2009.11.JNS091314
  121. Gainer JL, Sheehan JP, Larner JM, Jones DR. Trans sodium crocetinate with temozolomide and radiation therapy for glioblastoma multiforme. Journal of neurosurgery. Feb 2017;126(2):460-466. doi:10.3171/2016.3.JNS152693
  122. Whitten KM. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US). Identifier NCT00826930. Open-label Study to Determine the Effect of Trans Sodium Crocetinate (TSC) on Intra-tumoral Oxygen Concentration, Tolerability, and Pharmacokinetics of TSC in Post-operative Patients With High Grade Glioma (HGG). Last updated 8/12/2010. Accessed 12/22/2020. https://clinicaltrials.gov/ct2/show/NCT00826930.
  123. Diaz RJ, Ali S, Qadir MG, De La Fuente MI, Ivan ME, Komotar RJ. The role of bevacizumab in the treatment of glioblastoma. Journal of neuro-oncology. Jul 2017;133(3):455-467. doi:10.1007/s11060-017-2477-x
  124. Anthony C, Mladkova-Suchy N, Adamson DC. The evolving role of antiangiogenic therapies in glioblastoma multiforme: current clinical significance and future potential. Expert opinion on investigational drugs. 2019/09// 2019;28(9):787-797. doi:10.1080/13543784.2019.1650019
  125. Ahir BK, Engelhard HH, Lakka SS. Tumor Development and Angiogenesis in Adult Brain Tumor: Glioblastoma. Molecular neurobiology. May 2020;57(5):2461-2478. doi:10.1007/s12035-020-01892-8
  126. Tamura R, Tanaka T, Miyake K, Yoshida K, Sasaki H. Bevacizumab for malignant gliomas: current indications, mechanisms of action and resistance, and markers of response. Brain tumor pathology. Apr 2017;34(2):62-77. doi:10.1007/s10014-017-0284-x
  127. Narayana A, Yamada J, Berry S, et al. Intensity-modulated radiotherapy in high-grade gliomas: clinical and dosimetric results. International journal of radiation oncology, biology, physics. Mar 1 2006;64(3):892-7. doi:10.1016/j.ijrobp.2005.05.067
  128. Gzell C, Back M, Wheeler H, Bailey D, Foote M. Radiotherapy in Glioblastoma: the Past, the Present and the Future. Clinical oncology (Royal College of Radiologists (Great Britain)). Jan 2017;29(1):15-25. doi:10.1016/j.clon.2016.09.015
  129. Hottinger AF, Pacheco P, Stupp R. Tumor treating fields: a novel treatment modality and its use in brain tumors. Neuro Oncol. Oct 2016;18(10):1338-49. doi:10.1093/neuonc/now182
  130. Saria MG, Kesari S. Efficacy and Safety of Treating Glioblastoma With Tumor-Treating Fields Therapy. Clinical journal of oncology nursing. Oct 1 2016;20(5 Suppl):S9-S13. doi:10.1188/16.CJON.S1.9-13
  131. Lacouture ME, Anadkat MJ, Ballo MT, et al. Prevention and Management of Dermatologic Adverse Events Associated With Tumor Treating Fields in Patients With Glioblastoma. Frontiers Media S.A.; 2020.
  132. Stupp R, Taillibert S, Kanner A, et al. Effect of Tumor-Treating Fields Plus Maintenance Temozolomide vs Maintenance Temozolomide Alone on Survival in Patients With Glioblastoma: A Randomized Clinical Trial. JAMA. Dec 19 2017;318(23):2306-2316. doi:10.1001/jama.2017.18718
  133. Voloshin T, Kaynan N, Davidi S, et al. Tumor-treating fields (TTFields) induce immunogenic cell death resulting in enhanced antitumor efficacy when combined with anti-PD-1 therapy. Cancer immunology, immunotherapy : CII. Jul 2020;69(7):1191-1204. doi:10.1007/s00262-020-02534-7
  134. Seekatz B, Lukasczik M, Lohr M, et al. Screening for symptom burden and supportive needs of patients with glioblastoma and brain metastases and their caregivers in relation to their use of specialized palliative care. Supportive care in cancer : official journal of the Multinational Association of Supportive Care in Cancer. Sep 2017;25(9):2761-2770. doi:10.1007/s00520-017-3687-7
  135. Koekkoek JA, Chang S, Taphoorn MJ. Palliative care at the end-of-life in glioma patients. Handbook of clinical neurology. 2016;134:315-26. doi:10.1016/B978-0-12-802997-8.00019-0
  136. Finn OJ. A Believer's Overview of Cancer Immunosurveillance and Immunotherapy. J Immunol. Jan 15 2018;200(2):385-391. doi:10.4049/jimmunol.1701302
  137. Beatty GL, Gladney WL. Immune escape mechanisms as a guide for cancer immunotherapy. Clin Cancer Res. Feb 15 2015;21(4):687-92. doi:10.1158/1078-0432.CCR-14-1860
  138. Marcus A, Gowen BG, Thompson TW, et al. Recognition of tumors by the innate immune system and natural killer cells. Adv Immunol. 2014;122:91-128. doi:10.1016/B978-0-12-800267-4.00003-1
  139. Marin-Acevedo JA, Soyano AE, Dholaria B, Knutson KL, Lou Y. Cancer immunotherapy beyond immune checkpoint inhibitors. Journal of hematology & oncology. Jan 12 2018;11(1):8. doi:10.1186/s13045-017-0552-6
  140. Boussiotis VA, Charest A. Immunotherapies for malignant glioma. Oncogene. Mar 2018;37(9):1121-1141. doi:10.1038/s41388-017-0024-z
  141. Felthun J, Reddy R, McDonald KL. How immunotherapies are targeting the glioblastoma immune environment. Journal of clinical neuroscience : official journal of the Neurosurgical Society of Australasia. Jan 2018;47:20-27. doi:10.1016/j.jocn.2017.10.019
  142. Sampson JH, Gunn MD, Fecci PE, Ashley DM. Brain immunology and immunotherapy in brain tumours. Nat Rev Cancer. Jan 2020;20(1):12-25. doi:10.1038/s41568-019-0224-7
  143. Brown CE, Alizadeh D, Starr R, et al. Regression of Glioblastoma after Chimeric Antigen Receptor T-Cell Therapy. The New England journal of medicine. Dec 29 2016;375(26):2561-9. doi:10.1056/NEJMoa1610497
  144. Brown CE, Badie B, Barish ME, et al. Bioactivity and Safety of IL13Ralpha2-Redirected Chimeric Antigen Receptor CD8+ T Cells in Patients with Recurrent Glioblastoma. Clin Cancer Res. Sep 15 2015;21(18):4062-72. doi:10.1158/1078-0432.CCR-15-0428
  145. Brown CE, Aguilar B, Starr R, et al. Optimization of IL13Ralpha2-Targeted Chimeric Antigen Receptor T Cells for Improved Anti-tumor Efficacy against Glioblastoma. Molecular therapy : the journal of the American Society of Gene Therapy. Jan 3 2018;26(1):31-44. doi:10.1016/j.ymthe.2017.10.002
  146. Migliorini D, Dietrich PY, Stupp R, Linette GP, Posey AD, Jr., June CH. CAR T-Cell Therapies in Glioblastoma: A First Look. Clin Cancer Res. Feb 1 2018;24(3):535-540. doi:10.1158/1078-0432.CCR-17-2871
  147. Ahmed N, Brawley V, Hegde M, et al. HER2-Specific Chimeric Antigen Receptor-Modified Virus-Specific T Cells for Progressive Glioblastoma: A Phase 1 Dose-Escalation Trial. JAMA oncology. Aug 1 2017;3(8):1094-1101. doi:10.1001/jamaoncol.2017.0184
  148. Reardon DA, Brandes AA, Omuro A, et al. Effect of Nivolumab vs Bevacizumab in Patients With Recurrent Glioblastoma: The CheckMate 143 Phase 3 Randomized Clinical Trial. JAMA oncology. Jul 1 2020;6(7):1003-1010. doi:10.1001/jamaoncol.2020.1024
  149. Schalper KA, Rodriguez-Ruiz ME, Diez-Valle R, et al. Neoadjuvant nivolumab modifies the tumor immune microenvironment in resectable glioblastoma. Nat Med. Mar 2019;25(3):470-476. doi:10.1038/s41591-018-0339-5
  150. Smith C, Lineburg KE, Martins JP, et al. Autologous CMV-specific T cells are a safe adjuvant immunotherapy for primary glioblastoma multiforme. J Clin Invest. Nov 2 2020;130(11):6041-6053. doi:10.1172/JCI138649
  151. Brown MC, Gromeier M. Cytotoxic and immunogenic mechanisms of recombinant oncolytic poliovirus. Current opinion in virology. Aug 2015;13:81-5. doi:10.1016/j.coviro.2015.05.007
  152. Desjardins A, Sampson JH, Peters KB, et al. Phase I study of an Oncolytic polio/rhinovirus recombinant (PVSRIPO) against recurrent glioblastoma. Neuro-Oncology. 2013;15(suppl_3):iii107-iii107.
  153. Goetz C, Dobrikova E, Shveygert M, Dobrikov M, Gromeier M. Oncolytic poliovirus against malignant glioma. Future virology. Sep 2011;6(9):1045-1058. doi:10.2217/fvl.11.76
  154. Holl EK, Brown MC, Boczkowski D, et al. Recombinant oncolytic poliovirus, PVSRIPO, has potent cytotoxic and innate inflammatory effects, mediating therapy in human breast and prostate cancer xenograft models. Oncotarget. Nov 29 2016;7(48):79828-79841. doi:10.18632/oncotarget.12975
  155. Furuta T, Sabit H, Dong Y, et al. Biological basis and clinical study of glycogen synthase kinase- 3beta-targeted therapy by drug repositioning for glioblastoma. Oncotarget. Apr 4 2017;8(14):22811-22824. doi:10.18632/oncotarget.15206
  156. Denniston E, Crewdson H, Rucinsky N, et al. The Practical Consideration of Poliovirus as an Oncolytic Virotherapy. American journal of virology. 2016;5(1):1-7. doi:10.3844/ajvsp.2016.1.7
  157. Fong Y. Expediting Viral Therapies For Cancers To the Clinic. Molecular therapy : the journal of the American Society of Gene Therapy. Aug 2016;24(7):1161-2. doi:10.1038/mt.2016.128
  158. Desjardins A, Gromeier M, Herndon JE, 2nd, et al. Recurrent Glioblastoma Treated with Recombinant Poliovirus. The New England journal of medicine. Jul 12 2018;379(2):150-161. doi:10.1056/NEJMoa1716435
  159. Steiner HH, Bonsanto MM, Beckhove P, et al. Antitumor vaccination of patients with glioblastoma multiforme: a pilot study to assess feasibility, safety, and clinical benefit. J Clin Oncol. Nov 1 2004;22(21):4272-81. doi:10.1200/JCO.2004.09.038
  160. Huang B, Zhang H, Gu L, et al. Advances in Immunotherapy for Glioblastoma Multiforme. J Immunol Res. 2017;2017:3597613. doi:10.1155/2017/3597613
  161. Fenstermaker RA, Ciesielski MJ, Qiu J, et al. Clinical study of a survivin long peptide vaccine (SurVaxM) in patients with recurrent malignant glioma. Cancer immunology, immunotherapy : CII. Nov 2016;65(11):1339-1352. doi:10.1007/s00262-016-1890-x
  162. Saito T, Sugiyama K, Takeshima Y, et al. Prognostic implications of the subcellular localization of survivin in glioblastomas treated with radiotherapy plus concomitant and adjuvant temozolomide. Journal of neurosurgery. Mar 2018;128(3):679-684. doi:10.3171/2016.11.JNS162326
  163. Tang TK, Chiu SC, Lin CW, Su MJ, Liao MH. Induction of survivin inhibition, G(2)/M cell cycle arrest and autophagic on cell death in human malignant glioblastoma cells. The Chinese journal of physiology. Apr 30 2015;58(2):95-103. doi:10.4077/CJP.2015.BAC267
  164. Mellai M, Caldera V, Patrucco A, Annovazzi L, Schiffer D. Survivin expression in glioblastomas correlates with proliferation, but not with apoptosis. Anticancer research. Jan-Feb 2008;28(1A):109-18.
  165. Fenstermaker RA, Ciesielski MJ. Challenges in the development of a survivin vaccine (SurVaxM) for malignant glioma. Expert review of vaccines. Mar 2014;13(3):377-85. doi:10.1586/14760584.2014.881255
  166. Fenstermaker RA. A Phase II Study of the Safety and Efficacy of SVN53-67/M57-KLH (SurVaxM) in Survivin-Positive Newly Diagnosed Glioblastoma. U.S. National Library of Medicine. p. Identifier: NCT02455557-Identifier: NCT02455557.
  167. Ahluwalia M. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US). Identifier NCT04013672. Phase II Study of Pembrolizumab Plus SurVaxM for Glioblastoma at First Recurrence. Last updated 8/21/2020. Accessed 12/23/2020. https://www.clinicaltrials.gov/ct2/show/NCT04013672.
  168. Del Vecchio CA, Wong AJ. Rindopepimut, a 14-mer injectable peptide vaccine against EGFRvIII for the potential treatment of glioblastoma multiforme. Current opinion in molecular therapeutics. Dec 2010;12(6):741-54.
  169. Weller M, Butowski N, Tran DD, et al. Rindopepimut with temozolomide for patients with newly diagnosed, EGFRvIII-expressing glioblastoma (ACT IV): a randomised, double-blind, international phase 3 trial. The Lancet Oncology. Oct 2017;18(10):1373-1385. doi:10.1016/S1470-2045(17)30517-X
  170. Reardon DA, Desjardins A, Vredenburgh JJ, et al. Rindopepimut with Bevacizumab for Patients with Relapsed EGFRvIII-Expressing Glioblastoma (ReACT): Results of a Double-Blind Randomized Phase II Trial. Clin Cancer Res. Apr 1 2020;26(7):1586-1594. doi:10.1158/1078-0432.CCR-18-1140
  171. Chuang DF, Lin X. Targeted Therapies for the Treatment of Glioblastoma in Adults. Curr Oncol Rep. May 17 2019;21(7):61. doi:10.1007/s11912-019-0807-1
  172. Fujishiro T, Nonoguchi N, Pavliukov M, et al. 5-Aminolevulinic acid-mediated photodynamic therapy can target human glioma stem-like cells refractory to antineoplastic agents. Photodiagnosis Photodyn Ther. Dec 2018;24:58-68. doi:10.1016/j.pdpdt.2018.07.004
  173. Nakayama T, Otsuka S, Kobayashi T, et al. Dormant cancer cells accumulate high protoporphyrin IX levels and are sensitive to 5-aminolevulinic acid-based photodynamic therapy. Sci Rep. Nov 18 2016;6:36478. doi:10.1038/srep36478
  174. Ng WP, Liew BS, Idris Z, Rosman AK. Fluorescence-Guided versus Conventional Surgical Resection of High Grade Glioma: A Single-Centre, 7-Year, Comparative Effectiveness Study. Malays J Med Sci. Mar 2017;24(2):78-86. doi:10.21315/mjms2017.24.2.10
  175. Kast RE, Skuli N, Sardi I, et al. Augmentation of 5-Aminolevulinic Acid Treatment of Glioblastoma by Adding Ciprofloxacin, Deferiprone, 5-Fluorouracil and Febuxostat: The CAALA Regimen. Brain sciences. Nov 22 2018;8(12)doi:10.3390/brainsci8120203
  176. Kuo CJ, Leung LLK, Tirnauer JS. Overview of angiogenesis inhibitors. UpToDate. Updated 05/14/2020. Accessed 12/08/2020, https://www.uptodate.com/contents/overview-of-angiogenesis-inhibitors?search=glioblastoma%20regorafenib&source=search_result&selectedTitle=3~109&usage_type=default&display_rank=3
  177. Yarchoan M, Arnold SE. Repurposing diabetes drugs for brain insulin resistance in Alzheimer disease. Diabetes. Jul 2014;63(7):2253-61. doi:10.2337/db14-0287
  178. Seliger C, Meyer AL, Renner K, et al. Metformin inhibits proliferation and migration of glioblastoma cells independently of TGF-beta2. Cell cycle (Georgetown, Tex). Jul 2 2016;15(13):1755-66. doi:10.1080/15384101.2016.1186316
  179. Aldea MD, Petrushev B, Soritau O, et al. Metformin plus sorafenib highly impacts temozolomide resistant glioblastoma stem-like cells. Journal of BUON : official journal of the Balkan Union of Oncology. Apr-Jun 2014;19(2):502-11.
  180. Wurth R, Barbieri F, Florio T. New molecules and old drugs as emerging approaches to selectively target human glioblastoma cancer stem cells. Biomed Res Int. 2014;2014:126586. doi:10.1155/2014/126586
  181. Ferla R, Haspinger E, Surmacz E. Metformin inhibits leptin-induced growth and migration of glioblastoma cells. Oncology letters. Nov 2012;4(5):1077-1081. doi:10.3892/ol.2012.843
  182. Carmignani M, Volpe AR, Aldea M, et al. Glioblastoma stem cells: a new target for metformin and arsenic trioxide. Journal of biological regulators and homeostatic agents. Jan-Mar 2014;28(1):1-15.
  183. Gritti M, Wurth R, Angelini M, et al. Metformin repositioning as antitumoral agent: selective antiproliferative effects in human glioblastoma stem cells, via inhibition of CLIC1-mediated ion current. Oncotarget. Nov 30 2014;5(22):11252-68. doi:10.18632/oncotarget.2617
  184. Elmaci İ, Altinoz MA. A Metabolic Inhibitory Cocktail for Grave Cancers: Metformin, Pioglitazone and Lithium Combination in Treatment of Pancreatic Cancer and Glioblastoma Multiforme. Springer New York LLC; 2016. p. 573-618.
  185. Leidgens V, Proske J, Rauer L, et al. Stattic and metformin inhibit brain tumor initiating cells by reducing STAT3-phosphorylation. Oncotarget. Jan 31 2017;8(5):8250-8263. doi:10.18632/oncotarget.14159
  186. Sato A, Sunayama J, Okada M, et al. Glioma-initiating cell elimination by metformin activation of FOXO3 via AMPK. Stem cells translational medicine. Nov 2012;1(11):811-24. doi:10.5966/sctm.2012-0058
  187. Burkewitz K, Zhang Y, Mair WB. AMPK at the nexus of energetics and aging. Cell metabolism. Jul 1 2014;20(1):10-25. doi:10.1016/j.cmet.2014.03.002
  188. Riera CE, Merkwirth C, De Magalhaes Filho CD, Dillin A. Signaling Networks Determining Life Span. Annual review of biochemistry. Jun 2 2016;85:35-64. doi:10.1146/annurev-biochem-060815-014451
  189. Demaria M, Camporeale A, Poli V. STAT3 and metabolism: how many ways to use a single molecule? International journal of cancer Journal international du cancer. Nov 1 2014;135(9):1997-2003. doi:10.1002/ijc.28767
  190. Carpenter RL, Lo HW. STAT3 Target Genes Relevant to Human Cancers. Cancers. Apr 16 2014;6(2):897-925. doi:10.3390/cancers6020897
  191. Soritau O, Tomuleasa CI, Aldea M, et al. Metformin plus temozolomide-based chemotherapy as adjuvant treatment for WHO grade III and IV malignant gliomas. Journal of BUON: official journal of the Balkan Union of Oncology. 2011;16(2):282-289.
  192. Yang SH, Li S, Lu G, et al. Metformin treatment reduces temozolomide resistance of glioblastoma cells. Oncotarget. Nov 29 2016;7(48):78787-78803. doi:10.18632/oncotarget.12859
  193. Sesen J, Dahan P, Scotland SJ, et al. Metformin inhibits growth of human glioblastoma cells and enhances therapeutic response. PLoS One. 2015;10(4):e0123721. doi:10.1371/journal.pone.0123721
  194. Yu Z, Zhao G, Xie G, et al. Metformin and temozolomide act synergistically to inhibit growth of glioma cells and glioma stem cells in vitro and in vivo. Oncotarget. Oct 20 2015;6(32):32930-43. doi:10.18632/oncotarget.5405
  195. Lee JE, Lim JH, Hong YK, Yang SH. High-Dose Metformin Plus Temozolomide Shows Increased Anti-tumor Effects in Glioblastoma In Vitro and In Vivo Compared with Monotherapy. Cancer research and treatment : official journal of Korean Cancer Association. Oct 2018;50(4):1331-1342. doi:10.4143/crt.2017.466
  196. Adeberg S, Bernhardt D, Harrabi SB, et al. Metformin Enhanced in Vitro Radiosensitivity Associates with G2/M Cell Cycle Arrest and Elevated Adenosine-5'-monophosphate-activated Protein Kinase Levels in Glioblastoma. Radiology and oncology. Dec 2017;51(4):431-437. doi:10.1515/raon-2017-0042
  197. Zhao B, Wang X, Zheng J, Wang H, Liu J. Effects of metformin treatment on glioma-induced brain edema. Am J Transl Res. 2016;8(8):3351-63.
  198. Adeberg S, Bernhardt D, Ben Harrabi S, et al. Metformin influences progression in diabetic glioblastoma patients. Strahlentherapie und Onkologie : Organ der Deutschen Rontgengesellschaft [et al]. Dec 2015;191(12):928-35. doi:10.1007/s00066-015-0884-5
  199. Seliger C, Genbrugge E, Gorlia T, et al. Use of metformin and outcome of patients with newly diagnosed glioblastoma: Pooled analysis. International journal of cancer Journal international du cancer. Feb 1 2020;146(3):803-809. doi:10.1002/ijc.32337
  200. NIH. U.S. National Library of Medicine. ClinicalTrials.gov. Metformin: Glioblastoma. Accessed 6/4/2021, https://clinicaltrials.gov/ct2/results?term=metformin&cond=Glioblastoma&age_v=&gndr=&type=&rslt=&Search=Clear
  201. Agrawal S, Vamadevan P, Mazibuko N, et al. A New Method for Ethical and Efficient Evidence Generation for Off-Label Medication Use in Oncology (A Case Study in Glioblastoma). Frontiers in pharmacology. 2019;10(JUN):681. doi:10.3389/fphar.2019.00681
  202. Peng P, Wei W, Long C, Li J. Atorvastatin augments temozolomide's efficacy in glioblastoma via prenylation-dependent inhibition of Ras signaling. Biochemical and biophysical research communications. Jul 29 2017;489(3):293-298. doi:10.1016/j.bbrc.2017.05.147
  203. Petovari G, Hujber Z, Krencz I, et al. Targeting cellular metabolism using rapamycin and/or doxycycline enhances anti-tumour effects in human glioma cells. Cancer cell international. 2018;18(1):211. doi:10.1186/s12935-018-0710-0
  204. Skibinski CG, Williamson T, Riggins GJ. Mebendazole and radiation in combination increase survival through anticancer mechanisms in an intracranial rodent model of malignant meningioma. J Neurooncol. Dec 2018;140(3):529-538. doi:10.1007/s11060-018-03009-7
  205. Patil VM, Bhelekar A, Menon N, et al. Reverse swing-M, phase 1 study of repurposing mebendazole in recurrent high-grade glioma. Cancer Med. Jul 2020;9(13):4676-4685. doi:10.1002/cam4.3094
  206. Lubtow MM, Oerter S, Quader S, et al. In Vitro Blood-Brain Barrier Permeability and Cytotoxicity of an Atorvastatin-Loaded Nanoformulation Against Glioblastoma in 2D and 3D Models. Mol Pharm. Jun 1 2020;17(6):1835-1847. doi:10.1021/acs.molpharmaceut.9b01117
  207. Kalil AC, Freifeld AG, Lyden ER, Stoner JA. Valganciclovir for cytomegalovirus prevention in solid organ transplant patients: an evidence-based reassessment of safety and efficacy. PLoS One. 2009;4(5):e5512. doi:10.1371/journal.pone.0005512
  208. Stacpoole PW, Kurtz TL, Han Z, Langaee T. Role of dichloroacetate in the treatment of genetic mitochondrial diseases. Advanced drug delivery reviews. Oct-Nov 2008;60(13-14):1478-87. doi:10.1016/j.addr.2008.02.014
  209. Kankotia S, Stacpoole PW. Dichloroacetate and cancer: new home for an orphan drug? Biochim Biophys Acta. Dec 2014;1846(2):617-29. doi:10.1016/j.bbcan.2014.08.005
  210. Dunbar EM, Coats BS, Shroads AL, et al. Phase 1 trial of dichloroacetate (DCA) in adults with recurrent malignant brain tumors. Investigational new drugs. Jun 2014;32(3):452-64. doi:10.1007/s10637-013-0047-4
  211. Chu QS, Sangha R, Spratlin J, et al. A phase I open-labeled, single-arm, dose-escalation, study of dichloroacetate (DCA) in patients with advanced solid tumors. Investigational new drugs. Jun 2015;33(3):603-10. doi:10.1007/s10637-015-0221-y
  212. Michelakis ED, Sutendra G, Dromparis P, et al. Metabolic modulation of glioblastoma with dichloroacetate. Science translational medicine. May 12 2010;2(31):31ra34. doi:10.1126/scitranslmed.3000677
  213. Albatany M, Li A, Meakin S, Bartha R. Dichloroacetate induced intracellular acidification in glioblastoma: in vivo detection using AACID-CEST MRI at 9.4 Tesla. Journal of neuro-oncology. Jan 2018;136(2):255-262. doi:10.1007/s11060-017-2664-9
  214. James MO, Jahn SC, Zhong G, Smeltz MG, Hu Z, Stacpoole PW. Therapeutic applications of dichloroacetate and the role of glutathione transferase zeta-1. Pharmacology & therapeutics. Feb 2017;170:166-180. doi:10.1016/j.pharmthera.2016.10.018
  215. James MO, Stacpoole PW. Pharmacogenetic considerations with dichloroacetate dosing. Pharmacogenomics. May 2016;17(7):743-53. doi:10.2217/pgs-2015-0012
  216. Liu KH, Yang ST, Lin YK, et al. Fluoxetine, an antidepressant, suppresses glioblastoma by evoking AMPAR-mediated calcium-dependent apoptosis. Oncotarget. Mar 10 2015;6(7):5088-101. doi:10.18632/oncotarget.3243
  217. Song T, Li H, Tian Z, Xu C, Liu J, Guo Y. Disruption of NF-kappaB signaling by fluoxetine attenuates MGMT expression in glioma cells. OncoTargets and therapy. 2015;8:2199-208. doi:10.2147/OTT.S85948
  218. Bielecka-Wajdman AM, Lesiak M, Ludyga T, Sieron A, Obuchowicz E. Reversing glioma malignancy: a new look at the role of antidepressant drugs as adjuvant therapy for glioblastoma multiforme. Cancer chemotherapy and pharmacology. Jun 2017;79(6):1249-1256. doi:10.1007/s00280-017-3329-2
  219. Li J, Kim SG, Blenis J. Rapamycin: one drug, many effects. Cell metabolism. Mar 4 2014;19(3):373-9. doi:10.1016/j.cmet.2014.01.001
  220. Laplante M, Sabatini DM. mTOR signaling in growth control and disease. Cell. Apr 13 2012;149(2):274-93. doi:10.1016/j.cell.2012.03.017
  221. Jhanwar-Uniyal M, Amin AG, Cooper JB, Das K, Schmidt MH, Murali R. Discrete signaling mechanisms of mTORC1 and mTORC2: Connected yet apart in cellular and molecular aspects. Advances in biological regulation. May 2017;64:39-48. doi:10.1016/j.jbior.2016.12.001
  222. Masui K, Cavenee WK, Mischel PS. mTORC2 and Metabolic Reprogramming in GBM: at the Interface of Genetics and Environment. Brain Pathol. Nov 2015;25(6):755-9. doi:10.1111/bpa.12307
  223. Neil J, Shannon C, Mohan A, Laurent D, Murali R, Jhanwar-Uniyal M. ATP-site binding inhibitor effectively targets mTORC1 and mTORC2 complexes in glioblastoma. International journal of oncology. Mar 2016;48(3):1045-52. doi:10.3892/ijo.2015.3311
  224. Mendiburu-Elicabe M, Gil-Ranedo J, Izquierdo M. Efficacy of rapamycin against glioblastoma cancer stem cells. Clinical & translational oncology : official publication of the Federation of Spanish Oncology Societies and of the National Cancer Institute of Mexico. May 2014;16(5):495-502. doi:10.1007/s12094-013-1109-y
  225. Chheda MG, Wen PY, Hochberg FH, et al. Vandetanib plus sirolimus in adults with recurrent glioblastoma: results of a phase I and dose expansion cohort study. Journal of neuro-oncology. Feb 2015;121(3):627-34. doi:10.1007/s11060-014-1680-2
  226. Fan QW, Nicolaides TP, Weiss WA. Inhibiting 4EBP1 in Glioblastoma. Clin Cancer Res. Jan 1 2018;24(1):14-21. doi:10.1158/1078-0432.CCR-17-0042
  227. Cloughesy TF, Yoshimoto K, Nghiemphu P, et al. Antitumor activity of rapamycin in a Phase I trial for patients with recurrent PTEN-deficient glioblastoma. PLoS Med. Jan 22 2008;5(1):e8. doi:10.1371/journal.pmed.0050008
  228. Li X, Wu C, Chen N, et al. PI3K/Akt/mTOR signaling pathway and targeted therapy for glioblastoma. Oncotarget. May 31 2016;7(22):33440-50. doi:10.18632/oncotarget.7961
  229. Gulati N, Karsy M, Albert L, Murali R, Jhanwar-Uniyal M. Involvement of mTORC1 and mTORC2 in regulation of glioblastoma multiforme growth and motility. International journal of oncology. Oct 2009;35(4):731-40. doi:10.3892/ijo_00000386
  230. Luchman HA, Stechishin OD, Nguyen SA, Lun XQ, Cairncross JG, Weiss S. Dual mTORC1/2 blockade inhibits glioblastoma brain tumor initiating cells in vitro and in vivo and synergizes with temozolomide to increase orthotopic xenograft survival. Clin Cancer Res. Nov 15 2014;20(22):5756-67. doi:10.1158/1078-0432.CCR-13-3389
  231. Kahn J, Hayman TJ, Jamal M, et al. The mTORC1/mTORC2 inhibitor AZD2014 enhances the radiosensitivity of glioblastoma stem-like cells. Neuro Oncol. Jan 2014;16(1):29-37. doi:10.1093/neuonc/not139
  232. Atkins RJ, Stylli SS, Luwor RB, Kaye AH, Hovens CM. Glycogen synthase kinase-3beta (GSK-3beta) and its dysregulation in glioblastoma multiforme. Journal of clinical neuroscience : official journal of the Neurosurgical Society of Australasia. Sep 2013;20(9):1185-92. doi:10.1016/j.jocn.2013.02.003
  233. Kitabayashi T, Dong Y, Furuta T, et al. Identification of GSK3β inhibitor kenpaullone as a temozolomide enhancer against glioblastoma. Sci Rep. Jul 11 2019;9(1):10049. doi:10.1038/s41598-019-46454-8
  234. Pyko IV, Nakada M, Sabit H, et al. Glycogen synthase kinase 3β inhibition sensitizes human glioblastoma cells to temozolomide by affecting O6-methylguanine DNA methyltransferase promoter methylation via c-Myc signaling. Carcinogenesis. Oct 2013;34(10):2206-17. doi:10.1093/carcin/bgt182
  235. Niles RM. Recent advances in the use of vitamin A (retinoids) in the prevention and treatment of cancer. Nutrition (Burbank, Los Angeles County, Calif). Nov-Dec 2000;16(11-12):1084-9. doi:10.1016/s0899-9007(00)00436-6
  236. Uray IP, Dmitrovsky E, Brown PH. Retinoids and rexinoids in cancer prevention: from laboratory to clinic. Seminars in oncology. Feb 2016;43(1):49-64. doi:10.1053/j.seminoncol.2015.09.002
  237. Milani A, Basirnejad M, Shahbazi S, Bolhassani A. Carotenoids: biochemistry, pharmacology and treatment. Br J Pharmacol. Jun 2017;174(11):1290-1324. doi:10.1111/bph.13625
  238. Shapiro SS, Seiberg M, Cole CA. Vitamin A and its derivatives in experimental photocarcinogenesis: preventive effects and relevance to humans. J Drugs Dermatol. Apr 2013;12(4):458-63.
  239. Haque A, Banik NL, Ray SK. Emerging role of combination of all-trans retinoic acid and interferon-gamma as chemoimmunotherapy in the management of human glioblastoma. Neurochem Res. Dec 2007;32(12):2203-9. doi:10.1007/s11064-007-9420-z
  240. Yang D, Luo W, Wang J, et al. A novel controlled release formulation of the Pin1 inhibitor ATRA to improve liver cancer therapy by simultaneously blocking multiple cancer pathways. Journal of controlled release : official journal of the Controlled Release Society. Jan 10 2018;269:405-422. doi:10.1016/j.jconrel.2017.11.031
  241. Yin W, Song Y, Liu Q, Wu Y, He R. Topical treatment of all-trans retinoic acid inhibits murine melanoma partly by promoting CD8(+) T-cell immunity. Immunology. Oct 2017;152(2):287-297. doi:10.1111/imm.12768
  242. Friedman MD, Jeevan DS, Tobias M, Murali R, Jhanwar-Uniyal M. Targeting cancer stem cells in glioblastoma multiforme using mTOR inhibitors and the differentiating agent all-trans retinoic acid. Oncology reports. Oct 2013;30(4):1645-50. doi:10.3892/or.2013.2625
  243. Campos B, Wan F, Farhadi M, et al. Differentiation therapy exerts antitumor effects on stem-like glioma cells. Clin Cancer Res. May 15 2010;16(10):2715-28. doi:10.1158/1078-0432.CCR-09-1800
  244. Shi L, Li H, Zhan Y. All-trans retinoic acid enhances temozolomide-induced autophagy in human glioma cells U251 via targeting Keap1/Nrf2/ARE signaling pathway. Oncology letters. Sep 2017;14(3):2709-2714. doi:10.3892/ol.2017.6482
  245. Das A, Banik NL, Ray SK. Retinoids induced astrocytic differentiation with down regulation of telomerase activity and enhanced sensitivity to taxol for apoptosis in human glioblastoma T98G and U87MG cells. Journal of neuro-oncology. Mar 2008;87(1):9-22. doi:10.1007/s11060-007-9485-1
  246. Zhang C, Duvic M. Treatment of cutaneous T-cell lymphoma with retinoids. Dermatologic therapy. Sep-Oct 2006;19(5):264-71. doi:10.1111/j.1529-8019.2006.00083.x
  247. Heo JC, Jung TH, Lee S, et al. Effect of bexarotene on differentiation of glioblastoma multiforme compared with ATRA. Clinical & experimental metastasis. Jun 2016;33(5):417-29. doi:10.1007/s10585-016-9786-x
  248. Yung WK, Kyritsis AP, Gleason MJ, Levin VA. Treatment of recurrent malignant gliomas with high-dose 13-cis-retinoic acid. Clin Cancer Res. Dec 1996;2(12):1931-5.
  249. See SJ, Levin VA, Yung WK, Hess KR, Groves MD. 13-cis-retinoic acid in the treatment of recurrent glioblastoma multiforme. Neuro Oncol. Jul 2004;6(3):253-8. doi:10.1215/S1152851703000607
  250. Levin VA, Giglio P, Puduvalli VK, et al. Combination chemotherapy with 13-cis-retinoic acid and celecoxib in the treatment of glioblastoma multiforme. Journal of neuro-oncology. May 2006;78(1):85-90. doi:10.1007/s11060-005-9062-4
  251. Chen SE, Choi SS, Rogers JE, Lei X, De Groot JF. Isotretinoin maintenance therapy for glioblastoma: a retrospective review. J Oncol Pharm Pract. Apr 2014;20(2):112-9. doi:10.1177/1078155213483348
  252. Kast RE, Karpel-Massler G, Halatsch ME. CUSP9* treatment protocol for recurrent glioblastoma: aprepitant, artesunate, auranofin, captopril, celecoxib, disulfiram, itraconazole, ritonavir, sertraline augmenting continuous low dose temozolomide. Oncotarget. Sep 30 2014;5(18):8052-82. doi:10.18632/oncotarget.2408
  253. Skaga E, Skaga IØ, Grieg Z, Sandberg CJ, Langmoen IA, Vik-Mo EO. The efficacy of a coordinated pharmacological blockade in glioblastoma stem cells with nine repurposed drugs using the CUSP9 strategy. Journal of cancer research and clinical oncology. 2019;145(6):1495-1507.
  254. Picco S, Villegas L, Tonelli F, et al. Drug Repositioning for the Treatment of Glioma: Current State and Future Perspective. InTech Open; 2020:1-26.
  255. Toles M, Demark-Wahnefried W. Nutrition and the cancer survivor: evidence to guide oncology nursing practice. Seminars in oncology nursing. Aug 2008;24(3):171-9. doi:10.1016/j.soncn.2008.05.005
  256. Di Furia L, Rusciano MR, Leonardini L, et al. A Nutritional Approach to the Prevention of Cancer: from Assessment to Personalized Intervention. Translational medicine @ UniSa. Dec 2015;13:33-41.
  257. ACS. American Cancer Society. Benefits of good nutrition during cancer treatment. Updated 7/15/2019. Accessed 12/9/2020, https://www.cancer.org/treatment/survivorship-during-and-after-treatment/staying-active/nutrition/benefits.html
  258. Vanderwall C. Healthy Eating When You Have A Brain Tumor: Nutrition During Treatment. Ameican Brain Tumor Association; 2012.
  259. Filosa S, Di Meo F, Crispi S. Polyphenols-gut microbiota interplay and brain neuromodulation. Wolters Kluwer Medknow Publications; 2018. p. 2055-2059.
  260. Perrone L, Sampaolo S, Melone MAB. Bioactive Phenolic Compounds in the Modulation of Central and Peripheral Nervous System Cancers: Facts and Misdeeds. Cancers. Feb 15 2020;12(2)doi:10.3390/cancers12020454
  261. Levin GT, Greenwood KM, Singh F, Tsoi D, Newton RU. Exercise Improves Physical Function and Mental Health of Brain Cancer Survivors: Two Exploratory Case Studies. Integrative cancer therapies. Jun 2016;15(2):190-6. doi:10.1177/1534735415600068
  262. Winter SF, Loebel F, Dietrich J. Role of ketogenic metabolic therapy in malignant glioma: A systematic review. Critical reviews in oncology/hematology. Apr 2017;112:41-58. doi:10.1016/j.critrevonc.2017.02.016
  263. Paoli A, Bosco G, Camporesi EM, Mangar D. Ketosis, ketogenic diet and food intake control: a complex relationship. Frontiers in psychology. 2015;6:27. doi:10.3389/fpsyg.2015.00027
  264. Westman EC, Yancy WS, Jr., Mavropoulos JC, Marquart M, McDuffie JR. The effect of a low-carbohydrate, ketogenic diet versus a low-glycemic index diet on glycemic control in type 2 diabetes mellitus. Nutr Metab (Lond). Dec 19 2008;5:36. doi:10.1186/1743-7075-5-36
  265. Varshneya K, Carico C, Ortega A, Patil CG. The Efficacy of Ketogenic Diet and Associated Hypoglycemia as an Adjuvant Therapy for High-Grade Gliomas: A Review of the Literature. Cureus. Feb 2015;7(2):e251. doi:10.7759/cureus.251
  266. Klement RJ, Champ CE, Otto C, Kammerer U. Anti-Tumor Effects of Ketogenic Diets in Mice: A Meta-Analysis. PLoS One. 2016;11(5):e0155050. doi:10.1371/journal.pone.0155050
  267. Shelton LM, Huysentruyt LC, Mukherjee P, Seyfried TN. Calorie restriction as an anti-invasive therapy for malignant brain cancer in the VM mouse. ASN Neuro. Jul 23 2010;2(3):e00038. doi:10.1042/AN20100002
  268. Lussier DM, Woolf EC, Johnson JL, Brooks KS, Blattman JN, Scheck AC. Enhanced immunity in a mouse model of malignant glioma is mediated by a therapeutic ketogenic diet. BMC cancer. May 13 2016;16(1):310. doi:10.1186/s12885-016-2337-7
  269. Champ CE, Palmer JD, Volek JS, et al. Targeting metabolism with a ketogenic diet during the treatment of glioblastoma multiforme. Journal of neuro-oncology. Mar 2014;117(1):125-31. doi:10.1007/s11060-014-1362-0
  270. Meidenbauer JJ, Mukherjee P, Seyfried TN. The glucose ketone index calculator: a simple tool to monitor therapeutic efficacy for metabolic management of brain cancer. Nutr Metab (Lond). 2015;12(1):12. doi:10.1186/s12986-015-0009-2
  271. Klement RJ. Beneficial effects of ketogenic diets for cancer patients: a realist review with focus on evidence and confirmation. Medical oncology (Northwood, London, England). Aug 2017;34(8):132. doi:10.1007/s12032-017-0991-5
  272. Lin S. Ketogeic Diet Adjunctive to Salvage Chemotherapy for Recurrent Glioblastoma: a Pilot Study. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine. Identifier: NCT02939378. https://clinicaltrials.gov/ct2/show/NCT02939378?term=ketogenic&recrs=abdef&type=Intr&cond=glioblastoma&phase=01&rank=1. Last updated 10/20/2016. Accessed 1/16/2018.
  273. Klein P. Ketogenic Diet as Adjunctive Treatment in Refractory/End-stage Glioblastoma Multiforme: a Pilot Study. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine. Identifier: NCT01865162. https://clinicaltrials.gov/ct2/show/NCT01865162?term=ketogenic&recrs=abdef&type=Intr&cond=glioblastoma&phase=01&rank=2. Last updated 3/18/2016. Accessed 1/16/2018.
  274. Klein P. Ketogenic Diet Treatment Adjunctive to Radiation and Chemotherapy in Glioblastoma Multiforme: a Pilot Study. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine. Identifier: NCT02302235. https://clinicaltrials.gov/ct2/show/NCT02302235?term=ketogenic&recrs=abdef&type=Intr&cond=glioblastoma&phase=01&rank=3. Last updated 3/18/2016. Accessed 1/16/2018.
  275. Maroon JC, Seyfried TN, Donohue JP, Bost J. The role of metabolic therapy in treating glioblastoma multiforme. Surg Neurol Int. 2015;6:61. doi:10.4103/2152-7806.155259
  276. Seyfried TN, Marsh J, Shelton LM, Huysentruyt LC, Mukherjee P. Is the restricted ketogenic diet a viable alternative to the standard of care for managing malignant brain cancer? Epilepsy Res. Jul 2012;100(3):310-26. doi:10.1016/j.eplepsyres.2011.06.017
  277. Tan-Shalaby JL, Carrick J, Edinger K, et al. Modified Atkins diet in advanced malignancies - final results of a safety and feasibility trial within the Veterans Affairs Pittsburgh Healthcare System. Nutr Metab (Lond). 2016;13(1):52. doi:10.1186/s12986-016-0113-y
  278. Klement RJ. The influence of ketogenic therapy on the 5 R's of radiobiology. Int J Radiat Biol. Apr 2019;95(4):394-407. doi:10.1080/09553002.2017.1380330
  279. Seyfried TN, Shelton L, Arismendi-Morillo G, et al. Provocative Question: Should Ketogenic Metabolic Therapy Become the Standard of Care for Glioblastoma? Neurochem Res. Oct 2019;44(10):2392-2404. doi:10.1007/s11064-019-02795-4
  280. Weber DD, Aminzadeh-Gohari S, Tulipan J, Catalano L, Feichtinger RG, Kofler B. Ketogenic diet in the treatment of cancer – Where do we stand? : Elsevier GmbH; 2020. p. 102-121.
  281. Elsakka AMA, Bary MA, Abdelzaher E, et al. Management of Glioblastoma Multiforme in a Patient Treated With Ketogenic Metabolic Therapy and Modified Standard of Care: A 24-Month Follow-Up. Frontiers in nutrition. 2018;5:20. doi:10.3389/fnut.2018.00020
  282. Holick CN, Smith SG, Giovannucci E, Michaud DS. Coffee, tea, caffeine intake, and risk of adult glioma in three prospective cohort studies. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. Jan 2010;19(1):39-47. doi:10.1158/1055-9965.EPI-09-0732
  283. Guertin KA, Loftfield E, Boca SM, et al. Serum biomarkers of habitual coffee consumption may provide insight into the mechanism underlying the association between coffee consumption and colorectal cancer. Am J Clin Nutr. May 2015;101(5):1000-11. doi:10.3945/ajcn.114.096099
  284. Xue N, Zhou Q, Ji M, et al. Chlorogenic acid inhibits glioblastoma growth through repolarizating macrophage from M2 to M1 phenotype. Sci Rep. Jan 3 2017;7:39011. doi:10.1038/srep39011
  285. Belkaid A, Currie JC, Desgagnes J, Annabi B. The chemopreventive properties of chlorogenic acid reveal a potential new role for the microsomal glucose-6-phosphate translocase in brain tumor progression. Cancer cell international. Mar 27 2006;6:7. doi:10.1186/1475-2867-6-7
  286. Huber WW, Scharf G, Nagel G, Prustomersky S, Schulte-Hermann R, Kaina B. Coffee and its chemopreventive components Kahweol and Cafestol increase the activity of O6-methylguanine-DNA methyltransferase in rat liver--comparison with phase II xenobiotic metabolism. Mutation research. Jan 28 2003;522(1-2):57-68. doi:10.1016/s0027-5107(02)00264-6
  287. Fang MZ, Wang Y, Ai N, et al. Tea polyphenol (-)-epigallocatechin-3-gallate inhibits DNA methyltransferase and reactivates methylation-silenced genes in cancer cell lines. Cancer Res. Nov 15 2003;63(22):7563-70.
  288. Zhang Y, Wang SX, Ma JW, et al. EGCG inhibits properties of glioma stem-like cells and synergizes with temozolomide through downregulation of P-glycoprotein inhibition. Journal of neuro-oncology. Jan 2015;121(1):41-52. doi:10.1007/s11060-014-1604-1
  289. Chen TC, Wang W, Golden EB, et al. Green tea epigallocatechin gallate enhances therapeutic efficacy of temozolomide in orthotopic mouse glioblastoma models. Cancer letters. Mar 28 2011;302(2):100-8. doi:10.1016/j.canlet.2010.11.008
  290. Le CT, Leenders WPJ, Molenaar RJ, van Noorden CJF. Effects of the Green Tea Polyphenol Epigallocatechin-3-Gallate on Glioma: A Critical Evaluation of the Literature. Routledge; 2018. p. 317-333.
  291. Golden EB, Lam PY, Kardosh A, et al. Green tea polyphenols block the anticancer effects of bortezomib and other boronic acid-based proteasome inhibitors. Blood. Jun 4 2009;113(23):5927-37. doi:10.1182/blood-2008-07-171389
  292. Shah JJ, Kuhn DJ, Orlowski RZ. Bortezomib and EGCG: No green tea for you? : American Society of Hematology; 2009. p. 5695-5696.
  293. Armstrong TS, Shade MY, Breton G, et al. Sleep-wake disturbance in patients with brain tumors. Neuro Oncol. Mar 1 2017;19(3):323-335. doi:10.1093/neuonc/now119
  294. Zheng X, Pang B, Gu G, et al. Melatonin Inhibits Glioblastoma Stem-like cells through Suppression of EZH2-NOTCH1 Signaling Axis. International journal of biological sciences. 2017;13(2):245-253. doi:10.7150/ijbs.16818
  295. Zisapel N. New perspectives on the role of melatonin in human sleep, circadian rhythms and their regulation. Br J Pharmacol. Aug 2018;175(16):3190-3199. doi:10.1111/bph.14116
  296. Kurdi MS, Muthukalai SP. The Efficacy of Oral Melatonin in Improving Sleep in Cancer Patients with Insomnia: A Randomized Double-Blind Placebo-Controlled Study. Indian journal of palliative care. Jul-Sep 2016;22(3):295-300. doi:10.4103/0973-1075.185039
  297. Wade AG, Ford I, Crawford G, et al. Efficacy of prolonged release melatonin in insomnia patients aged 55-80 years: quality of sleep and next-day alertness outcomes. Current medical research and opinion. Oct 2007;23(10):2597-605. doi:10.1185/030079907X233098
  298. Wade AG, Ford I, Crawford G, et al. Nightly treatment of primary insomnia with prolonged release melatonin for 6 months: a randomized placebo controlled trial on age and endogenous melatonin as predictors of efficacy and safety. BMC Med. Aug 16 2010;8:51. doi:10.1186/1741-7015-8-51
  299. Favero G, Moretti E, Bonomini F, Reiter RJ, Fabrizio Rodella L, Rezzani R. Promising antineoplastic actions of melatonin. Frontiers Media S.A.; 2018.
  300. Chen X, Hao A, Li X, et al. Melatonin inhibits tumorigenicity of glioblastoma stem-like cells via the AKT-EZH2-STAT3 signaling axis. Journal of pineal research. Sep 2016;61(2):208-17. doi:10.1111/jpi.12341
  301. Zhang Y, Liu Q, Wang F, et al. Melatonin antagonizes hypoxia-mediated glioblastoma cell migration and invasion via inhibition of HIF-1alpha. Journal of pineal research. Sep 2013;55(2):121-30. doi:10.1111/jpi.12052
  302. Maitra S, Bhattacharya D, Das S, Bhattacharya S. Melatonin and its anti-glioma functions: a comprehensive review. Rev Neurosci. Jul 26 2019;30(5):527-541. doi:10.1515/revneuro-2018-0041
  303. Martin V, Sanchez-Sanchez AM, Herrera F, et al. Melatonin-induced methylation of the ABCG2/BCRP promoter as a novel mechanism to overcome multidrug resistance in brain tumour stem cells. British journal of cancer. May 28 2013;108(10):2005-12. doi:10.1038/bjc.2013.188
  304. Bojkova B, Kubatka P, Qaradakhi T, Zulli A, Kajo K. Melatonin May Increase Anticancer Potential of Pleiotropic Drugs. International journal of molecular sciences. Dec 6 2018;19(12):3910-3910. doi:10.3390/ijms19123910
  305. Anderson G, Reiter RJ. Glioblastoma: Role of Mitochondria N-acetylserotonin/Melatonin Ratio in Mediating Effects of miR-451 and Aryl Hydrocarbon Receptor and in Coordinating Wider Biochemical Changes. SAGE Publications Ltd; 2019.
  306. Lissoni P, Meregalli S, Nosetto L, et al. Increased survival time in brain glioblastomas by a radioneuroendocrine strategy with radiotherapy plus melatonin compared to radiotherapy alone. Oncology. Jan-Feb 1996;53(1):43-6. doi:10.1159/000227533
  307. Garcion E, Wion-Barbot N, Montero-Menei CN, Berger F, Wion D. New clues about vitamin D functions in the nervous system. Trends in Endocrinology & Metabolism. 2002;13(3):100-105. doi:10.1016/s1043-2760(01)00547-1
  308. Magrassi L, Adorni L, Montorfano G, et al. Vitamin D metabolites activate the sphingomyelin pathway and induce death of glioblastoma cells. Acta neurochirurgica. 1998;140(7):707-13; discussion 713-4. doi:10.1007/s007010050166
  309. Magrassi L, Butti G, Pezzotta S, Infuso L, Milanesi G. Effects of vitamin D and retinoic acid on human glioblastoma cell lines. Acta neurochirurgica. 1995;133(3-4):184-90. doi:10.1007/BF01420072
  310. Zhang Y, Dube C, Gibert M, Jr., et al. The p53 Pathway in Glioblastoma. Cancers. 2018;10(9):297. doi:10.3390/cancers10090297
  311. Cataldi S, Arcuri C, Lazzarini A, et al. Effect of 1alpha,25(OH)2 Vitamin D3 in Mutant P53 Glioblastoma Cells: Involvement of Neutral Sphingomyelinase1. Cancers. Oct 28 2020;12(11):3163-3163. doi:10.3390/cancers12113163
  312. Norlin M. Effects of vitamin D in the nervous system: Special focus on interaction with steroid hormone signalling and a possible role in the treatment of brain cancer. J Neuroendocrinol. Jan 2020;32(1):e12799. doi:10.1111/jne.12799
  313. Salomon DG, Fermento ME, Gandini NA, et al. Vitamin D receptor expression is associated with improved overall survival in human glioblastoma multiforme. Journal of neuro-oncology. May 2014;118(1):49-60. doi:10.1007/s11060-014-1416-3
  314. Bak DH, Kang SH, Choi DR, et al. Autophagy enhancement contributes to the synergistic effect of vitamin D in temozolomide-based glioblastoma chemotherapy. Experimental and therapeutic medicine. Jun 2016;11(6):2153-2162. doi:10.3892/etm.2016.3196
  315. Trouillas P, Honnorat J, Bret P, Jouvet A, Gerard JP. Redifferentiation therapy in brain tumors: long-lasting complete regression of glioblastomas and an anaplastic astrocytoma under long term 1-alpha-hydroxycholecalciferol. Journal of neuro-oncology. Jan 2001;51(1):57-66. doi:10.1023/a:1006437003352
  316. Tinggi U. Selenium: its role as antioxidant in human health. Environ Health Prev Med. Mar 2008;13(2):102-8. doi:10.1007/s12199-007-0019-4
  317. Yakubov E, Buchfelder M, Eyupoglu IY, Savaskan NE. Selenium action in neuro-oncology. Biol Trace Elem Res. Dec 2014;161(3):246-54. doi:10.1007/s12011-014-0111-8
  318. Philipov P, Tzatchev K. Selenium concentrations in serum of patients with cerebral and extracerebral tumors. Zentralblatt fur Neurochirurgie. 1988;49(4):344-7.
  319. Ertilav K, Naziroglu M, Ataizi ZS, Braidy N. Selenium Enhances the Apoptotic Efficacy of Docetaxel Through Activation of TRPM2 Channel in DBTRG Glioblastoma Cells. Neurotoxicity research. May 2019;35(4):797-808. doi:10.1007/s12640-019-0009-5
  320. Hazane-Puch F, Arnaud J, Trocme C, Faure P, Laporte F, Champelovier P. Sodium Selenite Decreased HDAC Activity, Cell Proliferation and Induced Apoptosis in Three Human Glioblastoma Cells. Anti-cancer agents in medicinal chemistry. 2016;16(4):490-500. doi:10.2174/1871520615666150819095426
  321. Zhu Z, Kimura M, Itokawa Y, Nakatsu S, Oda Y, Kikuchi H. Effect of selenium on malignant tumor cells of brain. Biol Trace Elem Res. Jul 1995;49(1):1-7. doi:10.1007/BF02788998
  322. Roomi MW, Ivanov V, Kalinovsky T, Niedzwiecki A, Rath M. Inhibition of glioma cell line A-172 MMP activity and cell invasion in vitro by a nutrient mixture. Medical oncology (Northwood, London, England). 2007;24(2):231-8. doi:10.1007/BF02698045
  323. Cheng Y, Sk UH, Zhang Y, et al. Rational incorporation of selenium into temozolomide elicits superior antitumor activity associated with both apoptotic and autophagic cell death. PLoS One. 2012;7(4):e35104. doi:10.1371/journal.pone.0035104
  324. Schneider H, Weller M. Boswellic acid activity against glioblastoma stem-like cells. Oncology letters. Jun 2016;11(6):4187-4192. doi:10.3892/ol.2016.4516
  325. Strowd RE, 3rd, Grossman SA. The Role of Glucose Modulation and Dietary Supplementation in Patients With Central Nervous System Tumors. Current treatment options in oncology. Aug 2015;16(8):36. doi:10.1007/s11864-015-0356-2
  326. Efferth T, Oesch F. Anti-inflammatory and anti-cancer activities of frankincense: Targets, treatments and toxicities. Academic Press.
  327. Roy NK, Deka A, Bordoloi D, et al. The potential role of boswellic acids in cancer prevention and treatment. Cancer letters. Jul 10 2016;377(1):74-86. doi:10.1016/j.canlet.2016.04.017
  328. Winking M, Sarikaya S, Rahmanian A, Jodicke A, Boker DK. Boswellic acids inhibit glioma growth: a new treatment option? Journal of neuro-oncology. 2000;46(2):97-103. doi:10.1023/a:1006387010528
  329. Li W, Ren L, Zheng X, et al. 3-O-Acetyl-11-keto- beta -boswellic acid ameliorated aberrant metabolic landscape and inhibited autophagy in glioblastoma. Acta pharmaceutica Sinica B. Feb 2020;10(2):301-312. doi:10.1016/j.apsb.2019.12.012
  330. Li W, Liu J, Fu W, et al. 3-O-acetyl-11-keto-beta-boswellic acid exerts anti-tumor effects in glioblastoma by arresting cell cycle at G2/M phase. Journal of experimental & clinical cancer research : CR. Jul 3 2018;37(1):132. doi:10.1186/s13046-018-0805-4
  331. Barbarisi M, Barbarisi A, De Sena G, et al. Boswellic acid has anti-inflammatory effects and enhances the anticancer activities of Temozolomide and Afatinib, an irreversible ErbB family blocker, in human glioblastoma cells. Phytother Res. Jun 2019;33(6):1670-1682. doi:10.1002/ptr.6354
  332. Conti S, Vexler A, Edry-Botzer L, et al. Combined acetyl-11-keto-beta-boswellic acid and radiation treatment inhibited glioblastoma tumor cells. PLoS One. 2018;13(7):e0198627. doi:10.1371/journal.pone.0198627
  333. Lin ZX. Glioma-related edema: new insight into molecular mechanisms and their clinical implications. Chinese journal of cancer. Jan 2013;32(1):49-52. doi:10.5732/cjc.012.10242
  334. Brandes AA, Tosoni A, Spagnolli F, et al. Disease progression or pseudoprogression after concomitant radiochemotherapy treatment: pitfalls in neurooncology. Neuro Oncol. Jun 2008;10(3):361-7. doi:10.1215/15228517-2008-008
  335. Streffer JR, Bitzer M, Schabet M, Dichgans J, Weller M. Response of radiochemotherapy-associated cerebral edema to a phytotherapeutic agent, H15. Neurology. May 8 2001;56(9):1219-21. doi:10.1212/wnl.56.9.1219
  336. Kirste S, Treier M, Wehrle SJ, et al. Boswellia serrata acts on cerebral edema in patients irradiated for brain tumors: a prospective, randomized, placebo-controlled, double-blind pilot trial. Cancer. Aug 15 2011;117(16):3788-95. doi:10.1002/cncr.25945
  337. Di Pierro F, Simonetti G, Petruzzi A, et al. A novel lecithin-based delivery form of Boswellic acids as complementary treatment of radiochemotherapy-induced cerebral edema in patients with glioblastoma multiforme: a longitudinal pilot experience. Journal of neurosurgical sciences. Jun 2019;63(3):286-291. doi:10.23736/S0390-5616.19.04662-9
  338. Sordillo LA, Sordillo PP, Helson L. Curcumin for the Treatment of Glioblastoma. Anticancer research. Dec 2015;35(12):6373-8.
  339. Klinger NV, Mittal S. Therapeutic Potential of Curcumin for the Treatment of Brain Tumors. Oxid Med Cell Longev. 2016;2016:9324085. doi:10.1155/2016/9324085
  340. Rodriguez GA, Shah AH, Gersey ZC, et al. Investigating the therapeutic role and molecular biology of curcumin as a treatment for glioblastoma. Therapeutic advances in medical oncology. Jul 2016;8(4):248-60. doi:10.1177/1758834016643518
  341. Shabaninejad Z, Pourhanifeh MH, Movahedpour A, et al. Therapeutic potentials of curcumin in the treatment of glioblstoma. European journal of medicinal chemistry. Feb 15 2020;188:112040. doi:10.1016/j.ejmech.2020.112040
  342. Wang Z, Liu F, Liao W, et al. Curcumin suppresses glioblastoma cell proliferation by p-AKT/mTOR pathway and increases the PTEN expression. Archives of biochemistry and biophysics. Aug 15 2020;689:108412. doi:10.1016/j.abb.2020.108412
  343. Fong D, Yeh A, Naftalovich R, Choi TH, Chan MM. Curcumin inhibits the side population (SP) phenotype of the rat C6 glioma cell line: towards targeting of cancer stem cells with phytochemicals. Cancer letters. Jul 1 2010;293(1):65-72. doi:10.1016/j.canlet.2009.12.018
  344. Zhuang W, Long L, Zheng B, et al. Curcumin promotes differentiation of glioma-initiating cells by inducing autophagy. Cancer science. Apr 2012;103(4):684-90. doi:10.1111/j.1349-7006.2011.02198.x
  345. Mukherjee S, Baidoo J, Fried A, et al. Curcumin changes the polarity of tumor-associated microglia and eliminates glioblastoma. International journal of cancer Journal international du cancer. Dec 15 2016;139(12):2838-2849. doi:10.1002/ijc.30398
  346. Perry MC, Demeule M, Regina A, Moumdjian R, Beliveau R. Curcumin inhibits tumor growth and angiogenesis in glioblastoma xenografts. Mol Nutr Food Res. Aug 2010;54(8):1192-201. doi:10.1002/mnfr.200900277
  347. Wang WH, Shen CY, Chien YC, et al. Validation of Enhancing Effects of Curcumin on Radiotherapy with F98/FGT Glioblastoma-Bearing Rat Model. International journal of molecular sciences. Jun 19 2020;21(12):4385-4385. doi:10.3390/ijms21124385
  348. Sminia P, van den Berg J, van Kootwijk A, Hageman E, Slotman BJ, Verbakel W. Experimental and clinical studies on radiation and curcumin in human glioma. Journal of cancer research and clinical oncology. Oct 28 2020:1-7. doi:10.1007/s00432-020-03432-2
  349. Mumtaz SM, Bhardwaj G, Goswami S, Tonk RK, Goyal RK, Pottoo FH. Management of Glioblastoma Multiforme by Phytochemicals: Applications of Nanoparticle Based Targeted Drug Delivery System. Current drug targets. Jul 26 2020;21doi:10.2174/1389450121666200727115454
  350. Shahcheraghi SH, Zangui M, Lotfi M, et al. Therapeutic Potential of Curcumin in the Treatment of Glioblastoma Multiforme. Curr Pharm Des. 2019;25(3):333-342. doi:10.2174/1381612825666190313123704
  351. Zhao J, Zhu J, Lv X, et al. Curcumin potentiates the potent antitumor activity of ACNU against glioblastoma by suppressing the PI3K/AKT and NF-kappaB/COX-2 signaling pathways. OncoTargets and therapy. 2017;10:5471-5482. doi:10.2147/OTT.S149708
  352. Xu M, Li G, Zhang H, et al. Sequential delivery of dual drugs with nanostructured lipid carriers for improving synergistic tumor treatment effect. Drug delivery. Dec 2020;27(1):983-995. doi:10.1080/10717544.2020.1785581
  353. Moussavi M, Assi K, Gomez-Munoz A, Salh B. Curcumin mediates ceramide generation via the de novo pathway in colon cancer cells. Carcinogenesis. Aug 2006;27(8):1636-44. doi:10.1093/carcin/bgi371
  354. Burgert A, Schlegel J, Becam J, et al. Characterization of Plasma Membrane Ceramides by Super-Resolution Microscopy. Angew Chem Int Ed Engl. May 22 2017;56(22):6131-6135. doi:10.1002/anie.201700570
  355. Stancevic B, Kolesnick R. Ceramide-rich platforms in transmembrane signaling. FEBS letters. May 3 2010;584(9):1728-40. doi:10.1016/j.febslet.2010.02.026
  356. Grammatikos G, Teichgraber V, Carpinteiro A, et al. Overexpression of acid sphingomyelinase sensitizes glioma cells to chemotherapy. Antioxid Redox Signal. Sep 2007;9(9):1449-56. doi:10.1089/ars.2007.1673
  357. Ashrafizadeh M, Mohammadinejad R, Farkhondeh T, Samarghandian S. Protective Effect of Resveratrol against Glioblastoma: A Review. Anti-cancer agents in medicinal chemistry. Sep 29 2020;20doi:10.2174/1871520620666200929151139
  358. Valentovic MA. Evaluation of Resveratrol in Cancer Patients and Experimental Models. Adv Cancer Res. 2018;137:171-188. doi:10.1016/bs.acr.2017.11.006
  359. Kiskova T, Kubatka P, Büsselberg D, Kassayova M. The plant-derived compound resveratrol in brain cancer: A review. MDPI AG; 2020.
  360. Zeng W, Zhang W, Lu F, Gao L, Gao G. Resveratrol attenuates MPP(+)-induced mitochondrial dysfunction and cell apoptosis via AKT/GSK-3beta pathway in SN4741 cells. Neurosci Lett. Jan 10 2017;637:50-56. doi:10.1016/j.neulet.2016.11.054
  361. Matsuno Y, Atsumi Y, Alauddin M, et al. Resveratrol and its Related Polyphenols Contribute to the Maintenance of Genome Stability. Sci Rep. Mar 25 2020;10(1):5388. doi:10.1038/s41598-020-62292-5
  362. Dionigi L, Ragonese F, Monarca L, et al. Focus on the Use of Resveratrol as an Adjuvant in Glioblastoma Therapy. Curr Pharm Des. 2020;26(18):2102-2108. doi:10.2174/1381612826666200401085634
  363. Mirzazadeh A, Kheirollahi M, Farashahi E, Sadeghian-Nodoushan F, Sheikhha MH, Aflatoonian B. Assessment Effects of Resveratrol on Human Telomerase Reverse Transcriptase Messenger Ribonucleic Acid Transcript in Human Glioblastoma. Advanced biomedical research. 2017;6:73. doi:10.4103/2277-9175.209047
  364. Clark PA, Bhattacharya S, Elmayan A, et al. Resveratrol targeting of AKT and p53 in glioblastoma and glioblastoma stem-like cells to suppress growth and infiltration. Journal of neurosurgery. May 2017;126(5):1448-1460. doi:10.3171/2016.1.JNS152077
  365. Song Y, Chen Y, Li Y, et al. Resveratrol Suppresses Epithelial-Mesenchymal Transition in GBM by Regulating Smad-Dependent Signaling. Biomed Res Int. 2019;2019(April 17):1321973. doi:10.1155/2019/1321973
  366. Jiao Y, Li H, Liu Y, et al. Resveratrol Inhibits the Invasion of Glioblastoma-Initiating Cells via Down-Regulation of the PI3K/Akt/NF-κB Signaling Pathway. Nutrients. 2015;7(6):4383-4402. doi:10.3390/nu7064383
  367. Li H, Liu Y, Jiao Y, et al. Resveratrol sensitizes glioblastoma-initiating cells to temozolomide by inducing cell apoptosis and promoting differentiation. Oncology reports. Jan 2016;35(1):343-51. doi:10.3892/or.2015.4346
  368. Yuan Y, Xue X, Guo RB, Sun XL, Hu G. Resveratrol enhances the antitumor effects of temozolomide in glioblastoma via ROS-dependent AMPK-TSC-mTOR signaling pathway. CNS neuroscience & therapeutics. Jul 2012;18(7):536-46. doi:10.1111/j.1755-5949.2012.00319.x
  369. Huang H, Lin H, Zhang X, Li J. Resveratrol reverses temozolomide resistance by downregulation of MGMT in T98G glioblastoma cells by the NF-kappaB-dependent pathway. Oncology reports. Jun 2012;27(6):2050-6. doi:10.3892/or.2012.1715
  370. Liu X, Wang Q, Liu B, et al. Genistein inhibits radiation-induced invasion and migration of glioblastoma cells by blocking the DNA-PKcs/Akt2/Rac1 signaling pathway. Radiother Oncol. Oct 28 2020;155:93-104. doi:10.1016/j.radonc.2020.10.026
  371. Wang L, Long L, Wang W, Liang Z. Resveratrol, a potential radiation sensitizer for glioma stem cells both in vitro and in vivo. Journal of pharmacological sciences. Dec 2015;129(4):216-25. doi:10.1016/j.jphs.2015.11.001
  372. Ozturk Y, Gunaydin C, Yalcin F, Naziroglu M, Braidy N. Resveratrol Enhances Apoptotic and Oxidant Effects of Paclitaxel through TRPM2 Channel Activation in DBTRG Glioblastoma Cells. Oxid Med Cell Longev. 2019;2019:4619865. doi:10.1155/2019/4619865
  373. Vidak M, Rozman D, Komel R. Effects of Flavonoids from Food and Dietary Supplements on Glial and Glioblastoma Multiforme Cells. Molecules (Basel, Switzerland). Oct 23 2015;20(10):19406-32. doi:10.3390/molecules201019406
  374. Natural Medicines Database. Quercetin. Professional monograph. https://naturalmedicines.therapeuticresearch.com/databases/food,-herbs-supplements/professional.aspx?productid=294
  375. Tavana E, Mollazadeh H, Mohtashami E, et al. Quercetin: A promising phytochemical for the treatment of glioblastoma multiforme. BioFactors (Oxford, England). May 2020;46(3):356-366. doi:10.1002/biof.1605
  376. Liu Y, Tang ZG, Yang JQ, et al. Low concentration of quercetin antagonizes the invasion and angiogenesis of human glioblastoma U251 cells. OncoTargets and therapy. 2017;10:4023-4028. doi:10.2147/OTT.S136821
  377. Kim H, Moon JY, Ahn KS, Cho SK. Quercetin induces mitochondrial mediated apoptosis and protective autophagy in human glioblastoma U373MG cells. Oxid Med Cell Longev. 2013;2013:596496. doi:10.1155/2013/596496
  378. Pan HC, Jiang Q, Yu Y, Mei JP, Cui YK, Zhao WJ. Quercetin promotes cell apoptosis and inhibits the expression of MMP-9 and fibronectin via the AKT and ERK signalling pathways in human glioma cells. Neurochemistry international. Jan 2015;80:60-71. doi:10.1016/j.neuint.2014.12.001
  379. Michaud-Levesque J, Bousquet-Gagnon N, Beliveau R. Quercetin abrogates IL-6/STAT3 signaling and inhibits glioblastoma cell line growth and migration. Experimental cell research. May 1 2012;318(8):925-35. doi:10.1016/j.yexcr.2012.02.017
  380. Sang DP, Li RJ, Lan Q. Quercetin sensitizes human glioblastoma cells to temozolomide in vitro via inhibition of Hsp27. Acta Pharmacol Sin. Jun 2014;35(6):832-8. doi:10.1038/aps.2014.22
  381. Pozsgai E, Bellyei S, Cseh A, et al. Quercetin increases the efficacy of glioblastoma treatment compared to standard chemoradiotherapy by the suppression of PI-3-kinase-Akt pathway. Nutrition and cancer. 2013;65(7):1059-66. doi:10.1080/01635581.2013.810291
  382. Jang E, Kim IY, Kim H, et al. Quercetin and chloroquine synergistically kill glioma cells by inducing organelle stress and disrupting Ca(2+) homeostasis. Biochemical pharmacology. Aug 2020;178:114098. doi:10.1016/j.bcp.2020.114098
  383. Siegelin MD, Habel A, Gaiser T. Epigalocatechin-3-gallate (EGCG) downregulates PEA15 and thereby augments TRAIL-mediated apoptosis in malignant glioma. Neurosci Lett. Dec 19 2008;448(1):161-5. doi:10.1016/j.neulet.2008.10.036
  384. Chu C, Deng J, Man Y, Qu Y. Green Tea Extracts Epigallocatechin-3-gallate for Different Treatments. Biomed Res Int. 2017;2017:5615647. doi:10.1155/2017/5615647
  385. Aggarwal V, Tuli HS, Tania M, et al. Molecular mechanisms of action of epigallocatechin gallate in cancer: Recent trends and advancement. Academic Press; 2020.
  386. Yokoyama S, Hirano H, Wakimaru N, Sarker KP, Kuratsu J. Inhibitory effect of epigallocatechin-gallate on brain tumor cell lines in vitro. Neuro Oncol. Jan 2001;3(1):22-8. doi:10.1093/neuonc/3.1.22
  387. Annabi B, Lachambre MP, Bousquet-Gagnon N, Page M, Gingras D, Beliveau R. Green tea polyphenol (-)-epigallocatechin 3-gallate inhibits MMP-2 secretion and MT1-MMP-driven migration in glioblastoma cells. Biochim Biophys Acta. Jan 30 2002;1542(1-3):209-20. doi:10.1016/s0167-4889(01)00187-2
  388. Bhattacharjee R, Devi A, Mishra S. Molecular docking and molecular dynamics studies reveal structural basis of inhibition and selectivity of inhibitors EGCG and OSU-03012 toward glucose regulated protein-78 (GRP78) overexpressed in glioblastoma. Journal of molecular modeling. Oct 2015;21(10):272. doi:10.1007/s00894-015-2801-3
  389. Xie CR, You CG, Zhang N, Sheng HS, Zheng XS. Epigallocatechin Gallate Preferentially Inhibits O(6)-Methylguanine DNA-Methyltransferase Expression in Glioblastoma Cells Rather than in Nontumor Glial Cells. Nutrition and cancer. Nov-Dec 2018;70(8):1339-1347. doi:10.1080/01635581.2018.1539189
  390. Sachinidis A, Seul C, Seewald S, Ahn H, Ko Y, Vetter H. Green tea compounds inhibit tyrosine phosphorylation of PDGF beta-receptor and transformation of A172 human glioblastoma. FEBS letters. Apr 7 2000;471(1):51-5. doi:10.1016/s0014-5793(00)01360-0
  391. Das A, Banik NL, Ray SK. Flavonoids activated caspases for apoptosis in human glioblastoma T98G and U87MG cells but not in human normal astrocytes. Cancer. Jan 1 2010;116(1):164-76. doi:10.1002/cncr.24699
  392. Udroiu I, Marinaccio J, Sgura A. Epigallocatechin-3-gallate induces telomere shortening and clastogenic damage in glioblastoma cells. Environmental and molecular mutagenesis. Oct 2019;60(8):683-692. doi:10.1002/em.22295
  393. Grube S, Ewald C, Kogler C, Lawson McLean A, Kalff R, Walter J. Achievable Central Nervous System Concentrations of the Green Tea Catechin EGCG Induce Stress in Glioblastoma Cells in Vitro. Nutrition and cancer. Oct 2018;70(7):1145-1158. doi:10.1080/01635581.2018.1495239
  394. Zhang J, Wang G, Mao Q, et al. Glutamate dehydrogenase (GDH) regulates bioenergetics and redox homeostasis in human glioma. Oncotarget. 02/26 2016;doi:10.18632/oncotarget.7657
  395. Peeters TH, Lenting K, Breukels V, et al. Isocitrate dehydrogenase 1-mutated cancers are sensitive to the green tea polyphenol epigallocatechin-3-gallate. Cancer Metab. 2019;7(1):4. doi:10.1186/s40170-019-0198-7
  396. Mani R, Natesan V. Chrysin: Sources, beneficial pharmacological activities, and molecular mechanism of action. Phytochemistry. Jan 2018;145:187-196. doi:10.1016/j.phytochem.2017.09.016
  397. Han JE, Lim PW, Na CM, et al. Inhibition of HIF1alpha and PDK Induces Cell Death of Glioblastoma Multiforme. Experimental neurobiology. Oct 2017;26(5):295-306. doi:10.5607/en.2017.26.5.295
  398. Noureddine H, Hage-Sleiman R, Wehbi B, et al. Chemical characterization and cytotoxic activity evaluation of Lebanese propolis. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie. Nov 2017;95:298-307. doi:10.1016/j.biopha.2017.08.067
  399. Santos BL, Oliveira MN, Coelho PL, et al. Flavonoids suppress human glioblastoma cell growth by inhibiting cell metabolism, migration, and by regulating extracellular matrix proteins and metalloproteinases expression. Chem Biol Interact. Dec 5 2015;242:123-38. doi:10.1016/j.cbi.2015.07.014
  400. Wang J, Wang H, Sun K, et al. Chrysin suppresses proliferation, migration, and invasion in glioblastoma cell lines via mediating the ERK/Nrf2 signaling pathway. Drug design, development and therapy. 2018;12(April):721-733. doi:10.2147/DDDT.S160020
  401. Markiewicz-Zukowska R, Borawska MH, Fiedorowicz A, Naliwajko SK, Sawicka D, Car H. Propolis changes the anticancer activity of temozolomide in U87MG human glioblastoma cell line. BMC Complement Altern Med. Feb 27 2013;13:50. doi:10.1186/1472-6882-13-50
  402. Stump TA, Santee BN, Williams LP, et al. The antiproliferative and apoptotic effects of apigenin on glioblastoma cells. The Journal of pharmacy and pharmacology. Jul 2017;69(7):907-916. doi:10.1111/jphp.12718
  403. Kim B, Jung N, Lee S, Sohng JK, Jung HJ. Apigenin Inhibits Cancer Stem Cell-Like Phenotypes in Human Glioblastoma Cells via Suppression of c-Met Signaling. Phytother Res. Nov 2016;30(11):1833-1840. doi:10.1002/ptr.5689
  404. Yi Y, Hsieh IY, Huang X, Li J, Zhao W. Glioblastoma Stem-Like Cells: Characteristics, Microenvironment, and Therapy. Frontiers in pharmacology. 2016;7:477. doi:10.3389/fphar.2016.00477
  405. Gursel DB, Shin BJ, Burkhardt JK, Kesavabhotla K, Schlaff CD, Boockvar JA. Glioblastoma stem-like cells-biology and therapeutic implications. Cancers. Jun 10 2011;3(2):2655-66. doi:10.3390/cancers3022655
  406. Freitas S, Costa S, Azevedo C, et al. Flavonoids inhibit angiogenic cytokine production by human glioma cells. Phytother Res. Jun 2011;25(6):916-21. doi:10.1002/ptr.3338
  407. Coelho PLC, Amparo JAO, da Silva AB, et al. Apigenin from Croton betulaster Mull restores the immune profile of microglia against glioma cells. Phytother Res. Dec 2019;33(12):3191-3202. doi:10.1002/ptr.6491
  408. Imran M, Aslam Gondal T, Atif M, et al. Apigenin as an anticancer agent. John Wiley and Sons Ltd; 2020. p. 1812-1828.
  409. Khani B, Mehrabian F, Khalesi E, Eshraghi A. Effect of soy phytoestrogen on metabolic and hormonal disturbance of women with polycystic ovary syndrome. Journal of research in medical sciences : the official journal of Isfahan University of Medical Sciences. Mar 2011;16(3):297-302.
  410. Carmichael SL, Gonzalez-Feliciano AG, Ma C, Shaw GM, Cogswell ME. Estimated dietary phytoestrogen intake and major food sources among women during the year before pregnancy. Nutr J. Oct 6 2011;10:105. doi:10.1186/1475-2891-10-105
  411. Cotterchio M, Boucher BA, Manno M, Gallinger S, Okey A, Harper P. Dietary phytoestrogen intake is associated with reduced colorectal cancer risk. J Nutr. Dec 2006;136(12):3046-53. doi:10.1093/jn/136.12.3046
  412. Honikl LS, Lammer F, Gempt J, Meyer B, Schlegel J, Delbridge C. High expression of estrogen receptor alpha and aromatase in glial tumor cells is associated with gender-independent survival benefits in glioblastoma patients. Journal of neuro-oncology. May 2020;147(3):567-575. doi:10.1007/s11060-020-03467-y
  413. Castracani CC, Longhitano L, Distefano A, et al. Role of 17β-Estradiol on Cell Proliferation and Mitochondrial Fitness in Glioblastoma Cells. Journal of oncology. 2020;2020:2314693. doi:10.1155/2020/2314693
  414. Zou S, Lang T, Liu X, Huang K, He X. Safety evaluation of genetically modified DAS-40278-9 maize in a subchronic rodent feeding study. Regulatory toxicology and pharmacology : RTP. Jul 2018;96:146-152. doi:10.1016/j.yrtph.2018.05.010
  415. Liu X, Liu K, Qin J, et al. C/EBPbeta promotes angiogenesis through secretion of IL-6, which is inhibited by genistein, in EGFRvIII-positive glioblastoma. International journal of cancer Journal international du cancer. Jun 1 2015;136(11):2524-34. doi:10.1002/ijc.29319
  416. Khaw AK, Yong JW, Kalthur G, Hande MP. Genistein induces growth arrest and suppresses telomerase activity in brain tumor cells. Genes, chromosomes & cancer. Oct 2012;51(10):961-74. doi:10.1002/gcc.21979
  417. Jafri MA, Ansari SA, Alqahtani MH, Shay JW. Roles of telomeres and telomerase in cancer, and advances in telomerase-targeted therapies. Genome medicine. Jun 20 2016;8(1):69. doi:10.1186/s13073-016-0324-x
  418. Chen X, Hao A, Li X, et al. Activation of JNK and p38 MAPK Mediated by ZDHHC17 Drives Glioblastoma Multiforme Development and Malignant Progression. Theranostics. 2020;10(3):998-1015. doi:10.7150/thno.40076
  419. Atefeh Z, Vahid C, Hasan N, Saeed A, Mahnaz H. Combination Treatment of Glioblastoma by Low-Dose Radiation and Genistein. Curr Radiopharm. 2016;9(3):258-263. doi:10.2174/1874471009666160813232031
  420. Siegelin MD, Gaiser T, Habel A, Siegelin Y. Daidzein overcomes TRAIL-resistance in malignant glioma cells by modulating the expression of the intrinsic apoptotic inhibitor, bcl-2. Neurosci Lett. May 1 2009;454(3):223-8. doi:10.1016/j.neulet.2009.03.031
  421. Yu C, Zhang P, Lou L, Wang Y. Perspectives Regarding the Role of Biochanin A in Humans. Frontiers in pharmacology. 2019;10:793-793. doi:10.3389/fphar.2019.00793
  422. Desai V, Jain A, Shaghaghi H, Summer R, Lai JCK, Bhushan A. Combination of Biochanin A and Temozolomide Impairs Tumor Growth by Modulating Cell Metabolism in Glioblastoma Multiforme. Anticancer research. Jan 2019;39(1):57-66. doi:10.21873/anticanres.13079
  423. Cheng YC, Hueng DY, Huang HY, Chen JY, Chen Y. Magnolol and honokiol exert a synergistic anti-tumor effect through autophagy and apoptosis in human glioblastomas. Oncotarget. May 17 2016;7(20):29116-30. doi:10.18632/oncotarget.8674
  424. Lin CJ, Chang YA, Lin YL, Liu SH, Chang CK, Chen RM. Preclinical effects of honokiol on treating glioblastoma multiforme via G1 phase arrest and cell apoptosis. Phytomedicine. May 15 2016;23(5):517-27. doi:10.1016/j.phymed.2016.02.021
  425. Lai IC, Shih PH, Yao CJ, et al. Elimination of cancer stem-like cells and potentiation of temozolomide sensitivity by Honokiol in glioblastoma multiforme cells. PLoS One. 2015;10(3):e0114830. doi:10.1371/journal.pone.0114830
  426. Zhang Y, Ren X, Shi M, et al. Downregulation of STAT3 and activation of MAPK are involved in the induction of apoptosis by HNK in glioblastoma cell line U87. Oncology reports. Nov 2014;32(5):2038-46. doi:10.3892/or.2014.3434
  427. Liang WZ, Chou CT, Chang HT, et al. The mechanism of honokiol-induced intracellular Ca(2+) rises and apoptosis in human glioblastoma cells. Chem Biol Interact. Sep 25 2014;221:13-23. doi:10.1016/j.cbi.2014.07.012
  428. Jeong JJ, Lee JH, Chang KC, Kim HJ. Honokiol exerts an anticancer effect in T98G human glioblastoma cells through the induction of apoptosis and the regulation of adhesion molecules. International journal of oncology. Oct 2012;41(4):1358-64. doi:10.3892/ijo.2012.1582
  429. Joo YN, Eun SY, Park SW, Lee JH, Chang KC, Kim HJ. Honokiol inhibits U87MG human glioblastoma cell invasion through endothelial cells by regulating membrane permeability and the epithelial-mesenchymal transition. International journal of oncology. Jan 2014;44(1):187-94. doi:10.3892/ijo.2013.2178
  430. Lin JW, Chen JT, Hong CY, et al. Honokiol traverses the blood-brain barrier and induces apoptosis of neuroblastoma cells via an intrinsic bax-mitochondrion-cytochrome c-caspase protease pathway. Neuro Oncol. Mar 2012;14(3):302-14. doi:10.1093/neuonc/nor217
  431. Zheng Z, Zhang J, Jiang J, et al. Remodeling tumor immune microenvironment (TIME) for glioma therapy using multi-targeting liposomal codelivery. Journal for immunotherapy of cancer. 2020;8(2):e000207. doi:10.1136/jitc-2019-000207
  432. Wu GJ, Yang ST, Chen RM. Major Contribution of Caspase-9 to Honokiol-Induced Apoptotic Insults to Human Drug-Resistant Glioblastoma Cells. Molecules (Basel, Switzerland). Mar 23 2020;25(6)doi:10.3390/molecules25061450
  433. Chio CC, Chen KY, Chang CK, et al. Improved effects of honokiol on temozolomide-induced autophagy and apoptosis of drug-sensitive and -tolerant glioma cells. BMC cancer. Apr 3 2018;18(1):379. doi:10.1186/s12885-018-4267-z
  434. Chio CC, Tai YT, Mohanraj M, Liu SH, Yang ST, Chen RM. Honokiol enhances temozolomide-induced apoptotic insults to malignant glioma cells via an intrinsic mitochondrion-dependent pathway. Phytomedicine. Oct 1 2018;49:41-51. doi:10.1016/j.phymed.2018.06.012
  435. Montecillo-Aguado M, Tirado-Rodriguez B, Tong Z, et al. Importance of the role of ω-3 and ω-6 polyunsaturated fatty acids in the progression of brain cancer. MDPI AG; 2020. p. 1-18.
  436. Kim S, Jing K, Shin S, et al. ω3-polyunsaturated fatty acids induce cell death through apoptosis and autophagy in glioblastoma cells: In vitro and in vivo. Oncology reports. 2018;39(1):239-246.
  437. Farago N, Feher LZ, Kitajka K, Das UN, Puskas LG. MicroRNA profile of polyunsaturated fatty acid treated glioma cells reveal apoptosis-specific expression changes. Lipids in health and disease. Sep 30 2011;10:173. doi:10.1186/1476-511X-10-173
  438. Das UN. From bench to the clinic: gamma-linolenic acid therapy of human gliomas. Prostaglandins Leukot Essent Fatty Acids. Jun 2004;70(6):539-52. doi:10.1016/j.plefa.2003.12.001
  439. Das UN. Gamma-linolenic acid therapy of human glioma-a review of in vitro, in vivo, and clinical studies. Med Sci Monit. Jul 2007;13(7):RA119-31.
  440. Das UN, Prasad VV, Reddy DR. Local application of gamma-linolenic acid in the treatment of human gliomas. Cancer letters. Aug 1 1995;94(2):147-55. doi:10.1016/0304-3835(95)03844-m
  441. Zou H, Zhu XX, Zhang GB, Ma Y, Wu Y, Huang DS. Silibinin: an old drug for hematological disorders. Oncotarget. Oct 24 2017;8(51):89307-89314. doi:10.18632/oncotarget.19153
  442. Ham J, Lim W, Bazer FW, Song G. Silibinin stimluates apoptosis by inducing generation of ROS and ER stress in human choriocarcinoma cells. J Cell Physiol. Feb 2018;233(2):1638-1649. doi:10.1002/jcp.26069
  443. Tuli HS, Mittal S, Aggarwal D, et al. Path of Silibinin from diet to medicine: A dietary polyphenolic flavonoid having potential anti-cancer therapeutic significance. Seminars in cancer biology. Oct 29 2020;doi:10.1016/j.semcancer.2020.09.014
  444. Bai ZL, Tay V, Guo SZ, Ren J, Shu MG. Silibinin Induced Human Glioblastoma Cell Apoptosis Concomitant with Autophagy through Simultaneous Inhibition of mTOR and YAP. Biomed Res Int. 2018;2018(March 26):6165192. doi:10.1155/2018/6165192
  445. Wang C, He C, Lu S, et al. Autophagy activated by silibinin contributes to glioma cell death via induction of oxidative stress-mediated BNIP3-dependent nuclear translocation of AIF. Cell Death Dis. Aug 14 2020;11(8):630. doi:10.1038/s41419-020-02866-3
  446. Momeny M, Malehmir M, Zakidizaji M, et al. Silibinin inhibits invasive properties of human glioblastoma U87MG cells through suppression of cathepsin B and nuclear factor kappa B-mediated induction of matrix metalloproteinase 9. Anti-cancer drugs. Mar 2010;21(3):252-60. doi:10.1097/cad.0b013e3283340cd7
  447. Chakrabarti M, Ray SK. Synergistic anti-tumor actions of luteolin and silibinin prevented cell migration and invasion and induced apoptosis in glioblastoma SNB19 cells and glioblastoma stem cells. Brain research. Dec 10 2015;1629:85-93. doi:10.1016/j.brainres.2015.10.010
  448. Chakrabarti M, Ray SK. Anti-tumor activities of luteolin and silibinin in glioblastoma cells: overexpression of miR-7-1-3p augmented luteolin and silibinin to inhibit autophagy and induce apoptosis in glioblastoma in vivo. Apoptosis. Mar 2016;21(3):312-28. doi:10.1007/s10495-015-1198-x
  449. Elhag R, Mazzio EA, Soliman KF. The effect of silibinin in enhancing toxicity of temozolomide and etoposide in p53 and PTEN-mutated resistant glioma cell lines. Anticancer research. Mar 2015;35(3):1263-9.
  450. Khairul I, Wang QQ, Jiang YH, Wang C, Naranmandura H. Metabolism, toxicity and anticancer activities of arsenic compounds. Oncotarget. Apr 4 2017;8(14):23905-23926. doi:10.18632/oncotarget.14733
  451. Lengfelder E, Hofmann WK, Nowak D. Impact of arsenic trioxide in the treatment of acute promyelocytic leukemia. Leukemia. Mar 2012;26(3):433-42. doi:10.1038/leu.2011.245
  452. Dizaji MZ, Malehmir M, Ghavamzadeh A, Alimoghaddam K, Ghaffari SH. Synergistic effects of arsenic trioxide and silibinin on apoptosis and invasion in human glioblastoma U87MG cell line. Neurochem Res. Feb 2012;37(2):370-80. doi:10.1007/s11064-011-0620-1
  453. Gulden M, Appel D, Syska M, Uecker S, Wages F, Seibert H. Chrysin and silibinin sensitize human glioblastoma cells for arsenic trioxide. Food and chemical toxicology : an international journal published for the British Industrial Biological Research Association. Jul 2017;105:486-497. doi:10.1016/j.fct.2017.04.035
  454. Comitato R, Ambra R, Virgili F. Tocotrienols: A Family of Molecules with Specific Biological Activities. Antioxidants (Basel, Switzerland). Nov 18 2017;6(4)doi:10.3390/antiox6040093
  455. Abubakar IB, Lim KH, Loh HS. Alkaloid extracts of Ficus species and palm oil-derived tocotrienols synergistically inhibit proliferation of human cancer cells. Natural product research. 2015;29(22):2137-40. doi:10.1080/14786419.2014.991927
  456. Aggarwal V, Kashyap D, Sak K, et al. Molecular Mechanisms of Action of Tocotrienols in Cancer: Recent Trends and Advancements. International journal of molecular sciences. Feb 2 2019;20(3)doi:10.3390/ijms20030656
  457. Lim SW, Loh HS, Ting KN, Bradshaw TD, Zeenathul NA. Cytotoxicity and apoptotic activities of alpha-, gamma- and delta-tocotrienol isomers on human cancer cells. BMC Complement Altern Med. Dec 6 2014;14:469. doi:10.1186/1472-6882-14-469
  458. Abubakar IB, Lim KH, Kam TS, Loh HS. Synergistic cytotoxic effects of combined delta-tocotrienol and jerantinine B on human brain and colon cancers. Journal of ethnopharmacology. May 26 2016;184:107-18. doi:10.1016/j.jep.2016.03.004
  459. Betti M, Minelli A, Canonico B, et al. Antiproliferative effects of tocopherols (vitamin E) on murine glioma C6 cells: homologue-specific control of PKC/ERK and cyclin signaling. Free radical biology & medicine. Aug 1 2006;41(3):464-72. doi:10.1016/j.freeradbiomed.2006.04.012
  460. Samandari E, Visarius T, Zingg JM, Azzi A. The effect of gamma-tocopherol on proliferation, integrin expression, adhesion, and migration of human glioma cells. Biochemical and biophysical research communications. Apr 21 2006;342(4):1329-33. doi:10.1016/j.bbrc.2006.02.110
  461. Ceci C, Lacal PM, Tentori L, De Martino MG, Miano R, Graziani G. Experimental Evidence of the Antitumor, Antimetastatic and Antiangiogenic Activity of Ellagic Acid. Nutrients. Nov 14 2018;10(11):1756-1756. doi:10.3390/nu10111756
  462. Wang D, Chen Q, Tan Y, Liu B, Liu C. Ellagic acid inhibits human glioblastoma growth in vitro and in vivo. Oncology reports. Feb 2017;37(2):1084-1092. doi:10.3892/or.2016.5331
  463. Wang D, Chen Q, Liu B, Li Y, Tan Y, Yang B. Ellagic acid inhibits proliferation and induces apoptosis in human glioblastoma cells. Acta cirurgica brasileira / Sociedade Brasileira para Desenvolvimento Pesquisa em Cirurgia. Feb 2016;31(2):143-9. doi:10.1590/S0102-865020160020000010
  464. Sitarek P, Skala E, Toma M, et al. A preliminary study of apoptosis induction in glioma cells via alteration of the Bax/Bcl-2-p53 axis by transformed and non-transformed root extracts of Leonurus sibiricus L. Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine. Jul 2016;37(7):8753-64. doi:10.1007/s13277-015-4714-2
  465. Cetin A, Biltekin B. Combining Ellagic Acid with Temozolomide Mediates the Cadherin Switch and Angiogenesis in a Glioblastoma Model. World neurosurgery. Dec 2019;132:e178-e184. doi:10.1016/j.wneu.2019.08.228
  466. Çetin A, Biltekin B, Degirmencioglu S. Ellagic Acid Enhances the Antitumor Efficacy of Bevacizumab in an In Vitro Glioblastoma Model. World neurosurgery. 2019/12// 2019;132:e59-e65. doi:10.1016/j.wneu.2019.08.257
  467. Hayakawa S, Ohishi T, Miyoshi N, Oishi Y, Nakamura Y, Isemura M. Anti-cancer effects of green tea epigallocatchin-3- gallate and coffee chlorogenic acid. MDPI AG; 2020.
  468. Saitou K, Ochiai R, Kozuma K, et al. Effect of Chlorogenic Acids on Cognitive Function: A Randomized, Double-Blind, Placebo-Controlled Trial. Nutrients. Sep 20 2018;10(10)doi:10.3390/nu10101337
  469. Sun C, Yu Y, Wang L, et al. Additive antiangiogenesis effect of ginsenoside Rg3 with low-dose metronomic temozolomide on rat glioma cells both in vivo and in vitro. Journal of experimental & clinical cancer research : CR. Feb 13 2016;35:32. doi:10.1186/s13046-015-0274-y
  470. Chen Z, Wei X, Shen L, Zhu H, Zheng X. 20(S)-ginsenoside-Rg3 reverses temozolomide resistance and restrains epithelial-mesenchymal transition progression in glioblastoma. Cancer science. Jan 2019;110(1):389-400. doi:10.1111/cas.13881
  471. Sin S, Kim SY, Kim SS. Chronic treatment with ginsenoside Rg3 induces Akt-dependent senescence in human glioma cells. International journal of oncology. Nov 2012;41(5):1669-74. doi:10.3892/ijo.2012.1604
  472. Nakhjavani M, Smith E, Townsend AR, Price TJ, Hardingham JE. Anti-Angiogenic Properties of Ginsenoside Rg3. NLM (Medline); 2020.
  473. Tunca B, Tezcan G, Cecener G, et al. Olea europaea leaf extract alters microRNA expression in human glioblastoma cells. Journal of cancer research and clinical oncology. Nov 2012;138(11):1831-44. doi:10.1007/s00432-012-1261-8
  474. Tezcan G, Tunca B, Bekar A, et al. Olea europaea leaf extract improves the treatment response of GBM stem cells by modulating miRNA expression. American journal of cancer research. 2014;4(5):572-90.
  475. Tezcan G, Tunca B, Demirci H, et al. Olea europaea Leaf Extract Improves the Efficacy of Temozolomide Therapy by Inducing MGMT Methylation and Reducing P53 Expression in Glioblastoma. Nutrition and cancer. Aug-Sep 2017;69(6):873-880. doi:10.1080/01635581.2017.1339810
  476. Tezcan G, Aksoy SA, Tunca B, et al. Oleuropein modulates glioblastoma miRNA pattern different from Olea europaea leaf extract. Human & experimental toxicology. Sep 2019;38(9):1102-1110. doi:10.1177/0960327119855123
  477. Majeed A, Muhammad Z, Ahmad H, Hayat SSS, Inayat N, Siyyar S. Nigella sativa L.: Uses in traditional and contemporary medicines–An overview. Acta Ecologica Sinica. 2020;
  478. Mollazadeh H, Afshari AR, Hosseinzadeh H. Review on the potential therapeutic roles of Nigella sativa in the treatment of patients with cancer: Involvement of apoptosis: - Black cumin and cancer. Korean Pharmacopuncture Institute; 2017. p. 158-172.
  479. Gali-Muhtasib H, Roessner A, Schneider-Stock R. Thymoquinone: a promising anti-cancer drug from natural sources. The international journal of biochemistry & cell biology. 2006;38(8):1249-53. doi:10.1016/j.biocel.2005.10.009
  480. Gomathinayagam R, Ha JH, Jayaraman M, Song YS, Isidoro C, Dhanasekaran DN. Chemopreventive and Anticancer Effects of Thymoquinone: Cellular and Molecular Targets. J Cancer Prev. Sep 30 2020;25(3):136-151. doi:10.15430/JCP.2020.25.3.136
  481. Racoma IO, Meisen WH, Wang QE, Kaur B, Wani AA. Thymoquinone inhibits autophagy and induces cathepsin-mediated, caspase-independent cell death in glioblastoma cells. PLoS One. 2013;8(9):e72882. doi:10.1371/journal.pone.0072882
  482. Krylova NG, Drobysh MS, Semenkova GN, Kulahava TA, Pinchuk SV, Shadyro OI. Cytotoxic and antiproliferative effects of thymoquinone on rat C6 glioma cells depend on oxidative stress. Molecular and cellular biochemistry. Dec 2019;462(1-2):195-206. doi:10.1007/s11010-019-03622-8
  483. Elmaci I, Altinoz MA. Thymoquinone: An edible redox-active quinone for the pharmacotherapy of neurodegenerative conditions and glial brain tumors. A short review. Elsevier Masson SAS; 2016. p. 635-640.
  484. Wieland A, Trageser D, Gogolok S, et al. Anticancer effects of niclosamide in human glioblastoma. Clin Cancer Res. Aug 1 2013;19(15):4124-36. doi:10.1158/1078-0432.CCR-12-2895
  485. Chen W, Mook RA, Premont RT, Wang J. Niclosamide: Beyond an antihelminthic drug. Elsevier Inc.; 2018. p. 89-96.
  486. Oh HC, Shim JK, Park J, et al. Combined effects of niclosamide and temozolomide against human glioblastoma tumorspheres. Journal of cancer research and clinical oncology. Nov 2020;146(11):2817-2828. doi:10.1007/s00432-020-03330-7
  487. Brodbelt A, Greenberg D, Winters T, Williams M, Vernon S, Collins VP. Glioblastoma in England: 2007-2011. Eur J Cancer. Mar 2015;51(4):533-542. doi:10.1016/j.ejca.2014.12.014
  488. Panwalkar P, Tamrazi B, Dang D, et al. Targeting integrated epigenetic and metabolic pathways in lethal childhood PFA ependymomas. Sci Transl Med. Oct 6 2021;13(614):eabc0497. doi:10.1126/scitranslmed.abc0497
  489. Archer Tenley C, Pomeroy Scott L. Posterior Fossa Ependymomas: A Tale of Two Subtypes. Cancer Cell. 2011/08/16/ 2011;20(2):133-134. doi:https://doi.org/10.1016/j.ccr.2011.08.003
  490. Marullo R, Castro M, Yomtoubian S, et al. The metabolic adaptation evoked by arginine enhances the effect of radiation in brain metastases. Science advances. Nov 5 2021;7(45):eabg1964. doi:10.1126/sciadv.abg1964
  491. Kim SH, Roszik J, Grimm EA, Ekmekcioglu S. Impact of l-Arginine Metabolism on Immune Response and Anticancer Immunotherapy. Frontiers in oncology. 2018;8:67. doi:10.3389/fonc.2018.00067
  492. Seltzer ES, Watters AK, MacKenzie D, Jr., Granat LM, Zhang D. Cannabidiol (CBD) as a Promising Anti-Cancer Drug. Cancers. Oct 30 2020;12(11)doi:10.3390/cancers12113203
  493. Sawtelle L, Holle LM. Use of Cannabis and Cannabinoids in Patients With Cancer. Ann Pharmacother. Jul 2021;55(7):870-890. doi:10.1177/1060028020965224
  494. Shahbazi F, Grandi V, Banerjee A, Trant JF. Cannabinoids and Cannabinoid Receptors: The Story so Far. iScience. Jul 24 2020;23(7):101301. doi:10.1016/j.isci.2020.101301
  495. Likar R, Kostenberger M, Nahler G. [Cannabidiol in cancer treatment]. Schmerz. Apr 2020;34(2):117-122. Cannabidiol bei Tumorerkrankungen. doi:10.1007/s00482-019-00438-9
  496. Baghban R, Roshangar L, Jahanban-Esfahlan R, et al. Tumor microenvironment complexity and therapeutic implications at a glance. Cell Commun Signal. Apr 7 2020;18(1):59. doi:10.1186/s12964-020-0530-4
  497. Jin MZ, Jin WL. The updated landscape of tumor microenvironment and drug repurposing. Signal transduction and targeted therapy. Aug 25 2020;5(1):166. doi:10.1038/s41392-020-00280-x
  498. Khodadadi H, Salles EL, Alptekin A, et al. Inhalant Cannabidiol Inhibits Glioblastoma Progression Through Regulation of Tumor Microenvironment. Cannabis Cannabinoid Res. Dec 16 2021;doi:10.1089/can.2021.0098
  499. Wang S, Wu J, Shen H, Wang J. The prognostic value of IDO expression in solid tumors: a systematic review and meta-analysis. BMC cancer. May 26 2020;20(1):471. doi:10.1186/s12885-020-06956-5