User:Drealle/Sandbox

Source: Wikipedia, the free encyclopedia.
Drealle/Sandbox
MRI
with contrast of a glioblastoma WHO grade IV in a 15-year-old boy
MRI
with contrast of a glioblastoma WHO grade IV in a 15-year-old boy
Glioblastoma (histology slide)

Glioblastoma multiforme (GBM) is the most common and aggressive type of primary

tumor
, accounting for 52% of all primary brain tumor cases and 20% of all intracranial tumors. Despite being the most prevalent form of primary brain tumor, GBMs occur in only 2–3 cases per 100,000 people in Europe and North America.

Treatment can involve

radiotherapy, and surgery, all of which are acknowledged as palliative measures, meaning that they do not provide a cure. Even with complete surgical resection
of the tumor, combined with the best available treatment, the survival rate for GBM remains very low. However, many advances in microsurgery techniques, radiotherapy and chemotherapy are slowly increasing the survival time of patients diagnosed with glioblastoma.

Glioblastoma multiform can be divided into three subgroups dependent on Karnofsky Performance Score (KPS), the age of the patient, and treatment:[1]

RPA class Definition Historical Median Survival Time Historical 1-Year Survival Historical 3-Year Survival Historical 5-Year Survival
III Age <50, KPS >= 90 17.1 months 70% 20% 14%
IV Age < 50, KPS < 90 11.2 months 46% 7% 4%
Age > 50, KPS >= 70, surgical removal with good neurologic function
V + VI Age >= 50, KPS >= 70, surgical removal with poor neurologic function 7.5 months 28% 1% 0%
Age >= 50, KPS >= 70, no surgical removal
Age >= 50, KPS < 70

Causes

GBM is more common in males, although the reason for this is not clear

electromagnetic fields[6], or viral infection[7]. There appears to be a small link between ionizing radiation and glioblastoma[8]
.

Pathogenesis

Glioblastomas multiforme are characterized by the presence of small areas of

astrocytomas
, post-mortem autopsies have revealed that most glioblastomas multiforme are not caused by previous lesions in the brain.

Unlike

Malignant cells carried in the CSF may spread to the spinal cord or cause meningeal gliomatosis. However, metastasis of GBM beyond the central nervous system is extremely rare. About 50% of GBM occupy more than one lobe of a hemisphere or are bilateral. Tumors of this type usually arise from the cerebrum and may exhibit the classic infiltrate across the corpus callosum, producing a butterfly (bilateral) glioma
.

The tumor may take on a variety of appearances, depending on the amount of hemorrhage,

lateral ventricle
is usually deformed, and both lateral and third ventricles may be displaced.

Symptoms

Although common symptoms of the disease include seizure, nausea and vomiting, headache, and hemiparesis, the single most prevalent symptom is a progressive memory, personality, or neurological deficit due to temporal and frontal lobe involvement. The kind of symptoms produced depends highly on the location of the tumor, more so than on its pathological properties. The tumor can start producing symptoms quickly, but occasionally is asymptomatic until it reaches an enormous size. See Symptoms section in:

Diagnosis

In early stages, when viewed with

MRI, glioblastoma may mimic more benign brain lesions[9]. Diagnosis of a suspected GBM on CT or MRI should rest on a stereotactic biopsy or by a craniotomy
, which can, at the same time, remove as much tumor as possible. Although the entire tumor can never be removed, in theory due to its multicentricity and diffuse character, partial resection ("debulking") can still prolong survival slightly.

Treatment

It is very difficult to treat glioblastoma due to several complicating factors:[10]

  • The tumor cells are very resistant to chemotherapy and other conventional therapies
  • The brain is susceptible to damage due to therapy
  • The brain has a very limited capacity to repair itself
  • Many drugs cannot cross the
    blood brain barrier
    to act on the tumor

Treatment of primary brain tumors and brain metastases consists of both symptomatic and palliative therapies.

Symptomatic therapy

Supportive treatment focuses on relieving symptoms and improving the patient’s neurologic function. The primary supportive agents are anticonvulsants and corticosteroids.

  • Historically, around ninety percent of patients with glioblastoma underwent anticonvulsant treatment, although is has been estimated that only approximately 40% of patients required this treatment. Recently, it has not been recommended that neurosurgeons administer anticonvulsants prophylactically, and should wait until a seizure occurs before prescribing this medication[11]. Those receiving phenytoin concurrent with radiation may have serious skin reactions such as erythema multiforme and Stevens–Johnson syndrome.
  • Corticosteroids, usually dexamethasone given 4 to 10 mg every 4 to 6 h, can reduce peritumoral edema (through rearrangement of the blood-brain barrier), diminishing mass effect and lowering intracranial pressure, with a decrease in headache or drowsiness.

Palliative therapy

Palliative treatment usually is conducted to improve quality of life and to achieve a longer survival time. It includes surgery, radiation therapy, and chemotherapy. A maximally feasible resection with maximal tumor-free margins ("debulking") is usually performed along with external beam radiation and chemotherapy.

Surgery

Surgery is the first stage of treatment of glioblastoma. An average GBM tumor contains 1011 cells, which is on average reduced to 109 cells after surgery. It is used to take a section for diagnosis, to remove some of the symptoms of a large mass pressing against the brain, to remove disease before secondary resistance to radiotherapy and chemotherapy, and to prolong survival.

The greater the extent of tumor removal, the longer the survival time. Removal of 98% or more of the tumor has been associated with a significantly longer median survival time than if less than 98% of the tumor is removed[12]. The chances of near-complete initial removal of the tumor can be greatly increased if the surgery is guided by a fluorescent dye known as 5-aminolevulinic acid[13].

Radiotherapy

On average,

radiotherapy after surgery can reduce the tumor size to 107 cells. Whole brain radiotherapy does not improve survival when compared to the more precise and targeted three-dimensional conformal radiotherapy[14]. A total radiation dose of 60-65 Gy has been found to be optimal for treatment [15]
.

Chemotherapy

The standard of care for glioblastoma includes chemotherapy during and after radiotherapy. On average, chemotherapy after surgery and radiotherapy can initially reduce the tumor size to 106 cells. The use of temozolomide both during radiotherapy and for six months post radiotherapy results in a significant increase in median survival with minimal additional toxicity[16]. This treatment regime is now standard for most cases of glioblastoma where the patient is not enrolled in a clinical trial[17][18].

Clinical trials

There are hundreds of

clinical trials
underway to assess the ability of new drugs and strategies to treat glioblastoma and extend the duration of survival. Below are some studies that have published promising results that have extended the survival of patients with GBM.

Pre-clinical studies
  • A possible therapy technique is to use viruses to attack the cancer. A modified herpes simplex virus has been engineered to invade tumor cells, and shows some efficacy in mice[19].
  • cell line, and rats injected with a human lung cancer cell and given dichloroacetate in their drinking water had smaller tumors than the rats that did not drink dichloroacetate[20]
    .
Phase I
  • Glioblastoma patients treated with motexafin gadolinium had a median survival time that was greater than a comparison group receiving standard care[21].
  • In recent studies, the antimalarial drug chloroquine has been shown to increase mid-term survival when given in combination with conventional therapy, although this difference did not reach statistical significance due to small sample size[22][23][24].
  • The use of injecting magnetic nanoparticles into the tumors of patients with recurrent GBM has been shown to be safe and feasible and resulted in promising survival rates.[25]. Promising survival rates have been observed in animal models[26], however, as yet there are not any results from efficacy studies in humans.
Phase II
Phase III

Recurrences

Long-term disease-free survival is unlikely, and the tumor will often reappear, usually within 2 cm of the original site, and 10% may develop new lesions at distant sites. More extensive surgery and intense local treatment after recurrence has been associated with improved survival.[34].

Prognosis

The median survival time from the time of diagnosis without any treatment is 3 months. Increasing age (> 60 years of age) carries a worse prognostic risk. Death is usually due to cerebral edema or increased intracranial pressure. One in twenty of glioblastoma patients survive for more than three years, and approximately one in 5,000 glioblastoma patients survive for decades[35]. Ben Williams has now survived glioblastoma for more than twelve years[36].

Survival of more than three years has been associated with younger age at diagnosis, a good initial KPS, and MGMT methylation [35]. A DNA test can be conducted on glioblastomas to determine whether or not the promoter of the MGMT gene is methylated. Patients with a methylated MGMT promoter have been associated with significantly greater long-term survival than patients with an unmethylated MGMT promoter[37]. This DNA characteristic is intrinsic to the patient and currently cannot be altered externally.

Long-term survival has also been associated with those patients who receive surgery, radiotherapy, and temozolomide chemotherapy[35]. However, much remains unknown about why some patients survive longer with glioblastoma.

References

  1. PMID 15758009. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link
    )
  2. .
  3. PMID 11319186. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link
    )
  4. PMID 14533876.{{cite journal}}: CS1 maint: multiple names: authors list (link
    )
  5. PMID 11150357. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link
    )
  6. PMID 9647903.{{cite journal}}: CS1 maint: multiple names: authors list (link
    )
  7. PMID 12798456. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link
    )
  8. .
  9. .
  10. ^ a b Lawson HC, Sampath P, Bohan E; et al. (2007). "Interstitial chemotherapy for malignant gliomas: the Johns Hopkins experience". Journal of Neuro-Oncology. 83 (1): 61–70.
    doi:10.1007/s11060-006-9303-1. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link) Cite error: The named reference "pmid12672279" was defined multiple times with different content (see the help page
    ).
  11. .
  12. PMID 11780887. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link
    )
  13. doi:10.1016/S1470-2045(06)70665-9. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link
    )
  14. doi:10.1016/j.ijrobp.2007.03.045. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: extra punctuation (link) CS1 maint: multiple names: authors list (link
    )
  15. PMID 1335044. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link
    )
  16. PMID 15758009. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link
    )
  17. doi:10.1017/S1748232105000054. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link
    )
  18. ^ "Temozolomide Plus Radiation Helps Brain Cancer - National Cancer Institute". Retrieved 2007-09-15.
  19. PMID 17479104
    .
  20. ^ Bonnet S, Archer S, Allalunis-Turner J; et al. (2007). "A Mitochondria-K+ Channel Axis Is Suppressed in Cancer and Its Normalization Promotes Apoptosis and Inhibits Cancer Growth". Cancer Cell. 11 (1): 37–51.
    doi:10.1016/j.ccr.2006.10.020. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link
    )
  21. PMID 17560737. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link
    )
  22. PMID 16520474.{{cite journal}}: CS1 maint: multiple names: authors list (link
    )
  23. PMID 17341043.{{cite journal}}: CS1 maint: multiple names: authors list (link
    )
  24. PMID 17350410.{{cite journal}}: CS1 maint: multiple names: authors list (link
    )
  25. ^ Maier-Hauff K, Rothe R, Scholz R; et al. (2007). "Intracranial thermotherapy using magnetic nanoparticles combined with external beam radiotherapy: results of a feasibility study on patients with glioblastoma multiforme". J Neurooncol. 81 (1).
    PMID 16773216. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link
    )
  26. PMID 16314937. {{cite journal}}: Explicit use of et al. in: |author= (help
    )
  27. ^ "TM601-001 – Phase I/II single-dose dose open label study in patients with recurrent glioma". Transmolecular, Inc. Retrieved 2008-02-04.
  28. ^ Reardon D, Akabani G, Friedman A; et al. "Outcome of Newly Diagnosed Patients with High-Grade Glioma Treated with Iodine-131 Murine Anti-Tenascin Monoclonal Antibody 81C6 via Surgically Created Resection Cavities in a Phase II Trial". Proc Am Soc Clin Oncol. 20. Retrieved 2008-02-04. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link)
  29. ^ Schlingensiepen KH, Schlingensiepen R, Steinbrecher A; et al. "Targeted tumor therapy with the TGF-beta2 antisense compound AP 12009". Cytokine Growth Factor Rev. 17 (1–7).
    PMID 16377233. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link
    )
  30. ^ Kleinberg L, Grossman SA, Carson K; et al. (2004). "Survival of Patients With Newly Diagnosed Glioblastoma Multiforme Treated With RSR13 and Radiotherapy: Results of a Phase II New Approaches to Brain Tumor Therapy CNS Consortium Safety and Efficacy Study". Journal of Clinical Oncology. 20 (14).
    doi:10.1215/S1152851703000127. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link
    )
  31. ^ Vredenburgh JJ, Desjardins A, Herndon JE; et al. (2007). "Bevacizumab Plus Irinotecan in Recurrent Glioblastoma Multiforme". Journal of Clinical Oncology. 25 (30).
    doi:10.1200/JCO.2007.12.2440. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link
    )
  32. PMID 15925768.{{cite journal}}: CS1 maint: multiple names: authors list (link
    )
  33. PMID 17926079.{{cite journal}}: CS1 maint: multiple names: authors list (link
    )
  34. doi:10.1016/j.critrevonc.2006.06.007.{{cite journal}}: CS1 maint: multiple names: authors list (link
    )
  35. ^
    doi:10.1093/brain/awm204. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link
    )
  36. ^ Williams, Ben (2006-8-28). "Brain Tumor Survivor Stories". Virtual Trials. Retrieved 2008-02-04. {{cite web}}: Check date values in: |date= (help)
  37. doi:10.1007/s11060-006-9292-0. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link
    )

Additional reading

External links