Jump to content
RemedySpot.com

Tumors and treatment

Rate this topic


Guest guest

Recommended Posts

Dear crew, whilst this is slanted towards older patients and malignancy, it

gives a good summary of different treatments available and current opinion

about effectiveness....also Nf gets a mention.

Site Map   Your Account   Support   About Us

Offerings:  Medscape.com   Charts   Mobile   Logician   CBSHealthwatch

From

Cancer Control: Journal of the Moffitt Cancer Center®

Brain Tumors in the Older Person

andra Flowers, MD, From the Department of Neurology at Hartford

Hospital, Hartford, Connecticut.

------------------------------------------------------------------------

Abstract

Background: The incidence of brain tumors is increasing rapidly,

particularly in the older population. Advances in molecular biology help to

explain differences in biologic behavior and response to therapy of brain

tumors in the elderly compared with younger patients. The number of elderly

patients who desire and receive therapy for brain tumors and are included in

clinical trials is increasing.

Methods: This article reviews the literature on the epidemiology, clinical

aspects, and therapy of brain tumors, with emphasis on the older patient

population.

Results: The increased incidence of brain tumors in the elderly is

principally due to the increasing number of people who comprise the older

population. Age and performance status are important independent prognostic

indicators, together with tumor histology. Surgery, radiation therapy, and

chemotherapy can benefit elderly patients with brain tumors with favorable

histologies, tumor location, and good performance status. The response rates

to available therapies are less favorable than in younger patients, and only

a small number of elderly patients are enrolled in clinical studies

addressing new treatment modalities.

Conclusions: Brain tumors in the elderly have specific characteristics that

determine their biologic behavior and response to therapy. There is a need

for clinical studies designed for treatment of brain tumors in older

patients, and requirements for rehabilitation and support systems for the

elderly need to be addressed. [Cancer Control; JMCC 7(6):523-538, 2000. ©

2000 Moffitt Cancer Center & Research Institute]

------------------------------------------------------------------------

Introduction

Brain tumors are primary or metastatic malignancies of the central nervous

system with considerable morbidity and mortality. The overall incidence of

brain tumors is increasing, with the highest increase noted in patients over

60 years of age. Until recently, these patients were managed with supportive

care only and were not considered eligible for clinical trials. The attitude

of the medical community toward offering treatments to elderly patients with

malignancies is changing, and more elderly patients with brain tumors are

now treated aggressively. [1] Advances in understanding the molecular

biology of brain tumors and the genetics of brain tumors in older patients

have resulted in treatments that are more effective or at least better

tolerated in this age group. The overall prognosis remains poor, however,

and the search for more effective therapies is ongoing.

Site Map   Your Account   Support   About Us

Offerings:  Medscape.com   Charts   Mobile   Logician   CBSHealthwatch

Brain Tumors in the Older Person continued...

[Cancer Control; JMCC 7(6):523-538, 2000. © 2000 Moffitt Cancer Center &

Research Institute]

------------------------------------------------------------------------

Epidemiology

Over the last 20 years, the overall incidence of cancer, including brain

cancer, has increased by more than 10%, as reported in the National Cancer

Institute statistics, with an average annual percentage change of

approximately 1%. [2-6] Between 1973 and 1985, there has been a dramatic

age-specific increase in the incidence of brain tumors. [2] The average

annual percentage increases in primary brain tumor incidence for ages 75-79,

80-84, and 85 and older were 7%, 20.4%, and 23.4%, respectively. [5-8] Since

1970, the incidence of primary brain tumors in people over the age of 70 has

increased sevenfold. [8] This trend continues in both the United States and

the industrialized European countries. [9-16] This increase in incidence

appears to be independent of diagnostic capabilities, although the

introduction of computed tomography (CT) scans in 1973, followed by magnetic

resonance imaging (MRI), allows for earlier and more accurate

diagnosis.[17,18] Comparisons between age-related mortality rates suggest

that increasing primary brain tumor mortality rates among the oldest age

groups are directly proportional to the increasing population size of these

age groups. [19-22] Malignant gliomas, particularly glioblastoma

multi-forme, are the most common primary brain tumors in the elderly.

The epidemiologic factors that have led to the increased incidence of brain

tumors in all age groups are not well defined. [23-25] The incidence of some

genetically transmitted diseases associated with brain tumors, such as

neurofibromatosis and the familial cancer syndromes (eg, Li-Fraumeni), has

not increased.[26,27] Also, there are no clearly established links between

the occurrence of brain tumors and environmental factors such as pesticides,

electromagnetic fields, and radiation exposure, except for higher risk for

meningiomas in patients who had previously received radiation therapy (RT)

to the head.[25,28] In some patients with a family history of malignancy,

there are abnormalities of tumor suppressor genes and overexpression of

oncogenes, which can be identified with molecular biology techniques.[26,27]

Age is a strong prognostic factor affecting survival.[29,30] An analysis

based on Surveillance, Epidemiology, and End Results (SEER) data for

1973-1991 shows that for patients aged 65 and older, there was no apparent

clinically significant improvement in survival rates for all tumor types

compared with significantly improved survival rates for younger patients

with anaplastic gliomas and medulloblastomas.[31,32] The 5-year survival

rate for patients with glioblastoma multiforme is approximately 20% in

patients less than 35 years of age, 10% in patients aged 35-54, and only 1%

in patients 55 years of age and older. [1] Similar age-related trends are

noted in patients with anaplastic astrocytomas (70%, 22%, and 15%,

respectively) (Fig 1). The age-based survival data parallel the survival

rates based on performance status, as measured by Karnofsky performance

score (KPS). Approximately 50% of patients with malignant gliomas over the

age of 55 are likely to have a KPS of less than 70 at diagnosis compared

with only 20% in the younger patients group (Fig 2). The performance status

is not the only determinant of survival in the elderly, but a low KPS

influences the type of treatment these patients are offered. [30]

Figure 1. (click image to zoom) Survival of patients with malignant gliomas

at 5 years by age (1980-1985 surveys). GBM = glioblastoma multiforme, AA =

anaplastic astrocytoma. From Flowers A. Brain tumors. In: Balducci L, Lyman

GH, Ershler WB, eds. Comprehensive Geriatric Oncology. Amsterdam, The

Netherlands: Harwood Academic; 1998:703-719. With permission from Gordon and

Breach Publishers.

Figure 2. (click image to zoom) Relationship of age and initial Karnofsky

performance score <70 in patients with malignant gliomas (1980-1985

surveys). GBM = glioblastoma multiforme, AA = anaplastic astrocytoma. From

Flowers A. Brain tumors. In: Balducci L, Lyman GH, Ershler WB, eds.

Comprehensive Geriatric Oncology. Amsterdam, The Netherlands: Harwood

Academic; 1998:703-719. With permission from Gordon and Breach Publishers.

continued... Site Map   Your Account   Support   About Us

Offerings:  Medscape.com   Charts   Mobile   Logician   CBSHealthwatch

Brain Tumors in the Older Person continued...

[Cancer Control; JMCC 7(6):523-538, 2000. © 2000 Moffitt Cancer Center &

Research Institute]

------------------------------------------------------------------------

Diagnosis

The diagnosis of brain tumors is based on clinical presentation, imaging

studies, and histology.[33,34] In the older population, intellectual decline

over a short period of time, gait disturbances, and short-term memory

deficits are clinical signs that may indicate the presence of a brain tumor

and must be differentiated from " normal " aging signs. [35]

Symptoms and Signs

The symptoms and signs are dependent on tumor location (Table 1). The

majority of tumors in the elderly are in the cerebral hemispheres. Headaches

and seizures are the most common symptoms at presentation. Headaches are

localized and persistent, and they increase in severity as the tumor grows

and exerts pressure. The seizures can be focal or generalized, and they may

have localizing value. The presence of focal neurologic deficits helps to

localize the lesion. The degree of neurologic compromise is an important

factor in deciding the therapeutic approach. Tumors in the anterior frontal

lobes, the anterior temporal lobes, or the base of the skull can grow to a

large size with few or no symptoms or with nonspecific symptoms often

ascribed to the aging process (eg, memory loss, personality changes, or some

gait difficulties). More than 60% of malignant gliomas arise in the frontal

and temporal lobes. The diagnosis of tumor can be suspected if the symptoms

develop over a short period of time (ie, less than 6 months). Nonspecific

cognitive and gait changes are also seen in patients with primary central

nervous system lymphomas. Unilateral hearing loss, vertigo, and mild face

weakness are symptoms caused by acoustic neuromas. Imaging studies help to

differentiate these from vertebrobasilar insufficiency.

Radiologic Diagnosis

Neuroimaging studies are valuable tools in localizing the lesion(s) and may

suggest the diagnosis and malignant character of a tumor. [36] Skull

radiographs can reveal abnormalities of the sella turcica, suggesting a

pituitary tumor or erosion of the bone as seen in patients with meningiomas,

as well as calcifications in low-grade astrocytomas, oligodendrogliomas, or

meningiomas. Cerebral angiograms help to distinguish tumors from vascular

malformations or aneurysms, and they also define the blood supply of the

tumor, thus assisting with the surgical management.

Contrast-enhanced CT and MRI scans of the brain are now the most utilized

imaging modalities. MRI scan of the brain is becoming the imaging modality

of choice for brain tumors. It allows visualization of the tumor in axial,

coronal, and sagittal planes, thereby providing a three-dimensional view of

the tumor and its relationship with the surrounding structures. MRI scans

have greater tissue contrast resolution than CT scans and allow the

visualization of not only very small lesions, but also lesions in the

temporal tip, in the inferior frontal lobe or posterior fossa, and at the

base of the skull. The paramagnetic substance gadolinium-diethylenetriamine

pentaacetic acid helps to define the intracranial lesions, to differentiate

neoplasms from other lesions, and to identify even subtle changes in the

appearance of a tumor during treatment. MRI scans with gadolinium are also

useful in diagnosing leptomeningeal metastases, which are seen with

increased frequency as brain tumor patients survive longer. Positron

emission tomography (PET) scans and single positron emission computed

tomography (SPECT) scans are less useful at diagnosis, but they can help to

distinguish tumor necrosis from radiation-induced necrosis in the follow-up

of tumors after therapy.[37,38] MR spectroscopy is still a research tool;

however, it has the potential to become a noninvasive diagnostic modality in

differentiating low-grade from anaplastic gliomas.

Pathologic Diagnosis

The pathologic examination of the tumor specimen on frozen section and

fixated material defines the type of tumor and the histologic grade. Brain

tumors can be primary or metastatic. Primary brain tumors are classified

histologically based on the World Health Organization classification. The

most common primary brain tumors are gliomas, which are classified according

to the cell type as astrocytic tumors, oligodendroglial tumors, or mixed

gliomas. The grade of malignancy is based on the cellularity, presence of

mitoses, vascular endothelial proliferation, and necrosis. [39] For an

accurate grading, the pathologist needs to know if the patient received RT

or chemotherapy prior to the surgical procedure. Tissue necrosis can be

caused by RT and chemotherapy as well as by some malignant tumors,

particularly glioblastoma multiforme.

Several classification schemes have been developed to grade malignant

gliomas (Table 2). [39-42] The histologic features are important

determinants of prognosis. Daumas-Duport et al [40] determined that the

length of postoperative survival is inversely proportional to the number of

histologic features of malignancy, such as nuclear atypia, mitoses, vascular

endothelial proliferation, and necrosis found in the tumor. [43] From a

practical standpoint, a three-tiered grading system is adequate for older

patients since even low-anaplastic tumors (grade II) tend to behave more

aggressively and need to be treated as anaplastic tumors rather than as

low-grade tumors. Gliomas that occur before the age of 10 and after the age

of 45 have a greater tendency to be undifferentiated and are associated with

more aggressive behavior and shorter postoperative survival. [44]

Oligodendrogliomas and mixed oligoastrocytomas carry a better prognosis.

[45] Determination of tumor cell proliferation pattern, using mitotic index

determination with bromodeoxyuridine (BUdR) or Ki-67 nuclear antigen or flow

cytometry, is being investigated as a way to define prognosis. [46-50]

In recent years, research has focused on defining the genetic alterations

and interactions among tumor-suppressor genes, oncogenes and their products,

growth factors, and enzyme systems.[48,49] The goal of this research is to

determine the mechanisms of onco-genesis, cell resistance, and repair

mechanisms and to develop new treatment modalities based on the molecular

biology data. The p53 tumor suppressor gene, found on chromosome 17p, is

frequently altered in gliomas as it is in systemic cancers. Tumors with a

high percentage of cells with mutated p53 gene grow rapid-ly and tend to

recur faster and be more resistant to therapy. Overexpression of p53 occurs

less often in patients over the age of 45.[51,52] Alterations on chromosome

17p are the most common seen in gliomas, even in low-grade gliomas. With a

higher grade of malignancy, other chromosomal alterations are seen on

chromosomes 9p, 19q, 4, 7, and 13. Alterations on chromosome 10 are seen in

50% of glioblastomas. The PTEN gene, located on 10q23, has been recently

identified as a putative tumor suppressor gene.[53,54] Mutations in the PTEN

gene have been found in high-grade adult gliomas and have a tendency to

occur in older patients. The loss of chromosome 10 has been directly

correlated with amplification of the epidermal growth factor receptor, which

can be targeted for therapeutic interventions.[53,54] Ploidy studies

indicate that for astrocytomas, survival is better for patients with

aneuploid tumors than with euploid tumors. No such correlation was found for

oligodendrogliomas. [49,52]

Meningiomas are more common in older patients (with a median age of 59) and

have a female predominance. The 5-year survival rate is 92% for patients

aged 45-74 and 70% for patients aged 75 and older.

Pituitary adenomas are also more common in the older population. In many

cases, asymptomatic microadenomas are found on scans of the brain done for

other reasons (eg, head trauma, headaches, or dizziness). Some patients

present with galactorrhea or with the physical changes of acromegaly.

Acoustic neuromas are benign tumors also seen in older patients. They should

be suspected in patients with unilateral hearing loss or vertigo that does

not resolve with medical treatment. Depending on the age of the patient, the

severity of symptoms, and the size of the tumor, management can be

conservative (with symptomatic treatment and follow-up with serial scans) or

more definitive (with surgery or stereotactic radiosurgery).

Differential Diagnosis

In older patients presenting with neurologic symptoms, the differential

diagnosis is primarily with cerebrovascular disease. Neuroimaging studies

can differentiate between tumor and stroke when the lesion does not respect

a vascular distribution. They can also help to differentiate between

hemorrhage due to hypertension and hemorrhage into a tumor. When the scan

reveals enhancing lesions, the differential diagnosis is between primary and

metastatic tumors. If the chest radiograph is normal, the most yielding

procedure will be a biopsy of one of the lesions for tissue diagnosis. In

this age group, infectious or vasculitic lesions are less common than in the

younger patients. younger patients.

Site Map   Your Account   Support   About Us

Offerings:  Medscape.com   Charts   Mobile   Logician   CBSHealthwatch

Brain Tumors in the Older Person continued...

[Cancer Control; JMCC 7(6):523-538, 2000. © 2000 Moffitt Cancer Center &

Research Institute]

Site Map   Your Account   Support   About Us

Offerings:  Medscape.com   Charts   Mobile   Logician   CBSHealthwatch

Brain Tumors in the Older Person continued...

[Cancer Control; JMCC 7(6):523-538, 2000. © 2000 Moffitt Cancer Center &

Research Institute]

------------------------------------------------------------------------

Treatment

The treatment of brain tumors is determined by the histologic type and the

location in the cranial cavity, as well as the patient's performance status,

neurologic status, age, and life expectancy as defined not only by the

neurologic deficits, but also by coexisting medical problems. [55-63] Benign

tumors such as meningiomas, acoustic neuromas, or pituitary adenomas can be

managed conservatively in older patients unless the symptoms warrant a more

aggressive approach. [64] In the elderly, treatment of primary brain tumors

raises particular challenges.

For gliomas, the conventional therapy involves surgery, RT, and

chemotherapy. [55-63] However, new treatment modalities are being developed,

as noted in Table 3. In the elderly, gliomas with a low-grade histology tend

to have a more aggressive biologic behavior and need to be treated with RT

and/or chemotherapy, as already mentioned.

Most primary brain tumors and metastatic tumors have surrounding vasogenic

edema, which contributes to the neurologic symptoms. The edema is controlled

with corticosteroids, diuretics or, in some cases, mannitol. The dosage of

corticosteroids is based on the amount of edema and mass effect. In patients

who undergo a wide resection of the tumor, corticosteroid use can be tapered

off relatively quickly. Side effects with long-term corticosteroid use are

gastric irritation, corticosteroid myopathy, Cushingoid appearance and, in

some patients, osteoporosis, depression, or corticosteroid psychosis. For

patients with diabetes who need to use corticosteroids, the blood glucose

must be monitored carefully, and insulin therapy may be needed to control

the hyperglycemia. In cases where corticosteroids are contraindicated (eg,

those with an active peptic ulcer, heart failure, or uncontrolled diabetes),

diuretics such as acetazolamide or furosemide can decrease the edema.

Surgery

Surgery is the first therapeutic intervention for brain tumors, with the

goal of obtaining tissue for diagnosis and, whenever possible, debulking the

tumor to relieve the pressure and bring about rapid clinical improvement.

[64] In the elderly, surgery is considered to carry a higher risk of

morbidity and mortality compared with younger patients.

When feasible, complete resection of the tumor has been shown to

significantly increase the rate of survival by improving the patient's

performance status and by providing cytoreduction, with a better chance of

response to subsequent therapy. Outcome studies also have shown that

patients who undergo resection have a better quality of life and are less

likely to become depressed than patients who undergo only biopsy. [65-67]

The survival advantage is particularly significant for anaplastic

astrocytomas. The 5-year survival rates are 50% in patients with

astrocytomas who had a total resection but only 20% in patients who had a

biopsy only. [65] For patients with unresectable lesions or with associated

significant medical problems, a stereotactic biopsy for tissue diagnosis is

sufficient. The most important prognostic factor remains the extent of

resection. The postoperative residual tumor volume (determined on enhanced

CT or MRI scans) correlates inversely with survival. [68,69]

Reoperation for recurrent or progressing tumors must be considered on a

case-by-case basis, depending on the tumor type, expected survival, KPS,

patient age, and plans for further therapy.[70,71] Age is a factor that can

influence the outcome. In one study, survival after reoperation for

recurrent gliomas was 57 weeks for patients younger than 40 years but only

36 weeks for older patients. [72] Other authors found a correlation between

age and overall survival from diagnosis but no difference after reoperation.

[66]

Perfected neurosurgical techniques such as computer-assisted minimal-access

surgery have reduced the morbidity associated with open craniotomies and

have shortened the length of hospital stay. These improvements have made

such interventions safer and more acceptable for elderly patients with

resectable tumors. [73]

Radiation Therapy

For malignant brain tumors, RT is considered to be the standard treatment.

[74-79] For malignant gliomas, RT at doses of 50-60 Gy increases survival

compared with surgery alone. [74] RT is also the treatment of choice for

low-grade gliomas in elderly patients. [75] For malignant gliomas, RT is

delivered to the area of tumor as visualized on CT or MRI scan plus an

additional 3-cm margin. [76] New software allows three-dimensional RT

planning. The total dose of radiation is delivered over a span of 30-33 days

in daily fractions of 1.6-2.0 Gy. Age is again an important prognostic

factor. In one study, the survival rate at 18 months was 64% in patients

younger than age 40 but only 8% in patients older than age 60. [78,79] The

performance status is an independent variable. The survival rate at 18

months for patients with an initial KPS of 70 or above is 34% compared with

13% for patients with a KPS score of 60 or below. [80]

The results of conventional fractionation RT have been disappointing in

terms of providing a cure or at least long-term survival in patients with

malignant gliomas. Reasons for failure are related to tumor cell resistance

(particularly in hypoxic areas of the tumor), the presence of repair

mechanisms, and the pattern of spread of these tumors along white matter

tracts out-side the radiation field. [81] Several different methods are

under investigation to enhance radiosensitivity, provide protection to the

normal brain tissue, and deliver higher doses of radiation to the tumor.

Investigational Radiation Therapy Methods

Hyperfractionation schedules allow the RT dose to be delivered in two or

three daily treatments, while hypofractionation schedules use a once-weekly

treatment. These modified RT schedules allow for either smaller doses

administered at shorter time intervals so larger total doses can be used

with less toxicity to the normal brain, or larger doses administered to

reduce the morbidity related to RT.

Hypofractionation schedules in which RT is delivered in weekly doses of

5.0-6.5 Gy over 6 weeks to a total dose of 36-39 Gy, together with

administration of cisplatin or carboplatin as radiosensitizers, have been

used for patients with a poor performance status (KPS of 60 or below). This

approach was well tolerated and improved both performance status and

survival in some of the patients. [82,83]

In accelerated fractionation schedules, the conventional dose of radiation

is delivered in two or three daily fractions. The rationale is that

shortened treatment time would improve the therapeutic ratio, with greater

tumor control. Drugs such as carboplatin or BUdR are used as

radiosensitizers. The toxicities in these studies are related to the

myelosuppressive effect of the cytotoxic agents -- skin rash with BUdR.

These schedules are well tolerated, even by older patients. Accelerated

fractionation has been used for hospitalized patients with poor performance

status in order to shorten the duration of treatment. [84]

Hypoxic cell sensitizers such as misonidazole or lonidamine in combination

with RT were promising in experimental studies. However, in randomized

clinical trials, these agents showed no benefit in survival.

Halogenated pyrimidine analogues such as BUdR or iododeoxyuridine (IUdR) are

incorporated into rapidly dividing cells and act as radiosensitizers. Phase

II studies noted an increase in survival over conventional RT. Other studies

have investigated the radiosensitizing effect of some chemotherapeutic

agents including hydroxyurea, vincristine, and BCNU (carmustine). RSR13, an

allosteric modifier of hemoglobin, is a novel radiosensitizer that binds

covalently to hemoglobin and reduces oxygen-binding affinity, thus

increasing oxygen release in the capillaries. RSR13 is now undergoing

clinical trials. The difference in survival compared with conventional RT is

not significant. Patients under the age of 60 seem to benefit most. [85]

Stereotactic radiosurgery is a noninvasive technique that allows delivery of

high-dose single fractions of radiation to small, well-circumscribed tumors.

The treatment is safe and effective, and because it is done in one single

dose or a few fractionated doses in an out-patient setting, stereotactic

radiosurgery is convenient for the patient and cost effective.[86,87] The

morbidity associated with this approach is primarily related to increased

peritumoral edema, which can be easily controlled with corticosteroids. To

date, no cognitive deficits have been described in patients who have

received radiosurgery without conventional RT. For treatment of malignant

gliomas, stereotactic radio-surgery is used as adjuvant therapy to external

beam RT. Because of the infiltrative pattern of growth of these tumors, this

technique cannot be used as the sole radiation modality. [88] Radiosurgery

can also be administered in fractionated doses. [86]

Interstitial RT (brachytherapy) is a more invasive way of delivering

high-dose radiation to a tumor while limiting the dose to the surrounding

brain. A dose of 60 Gy or more can be delivered even after 60 Gy of external

beam RT. Brachytherapy refers to treatment with radiation sources placed

directly into the tumor mass or adjacent to tumors (eg, in the surgical

cavity). The most commonly used isotopes for brachytherapy are 192Ir and

125I. Although more invasive than stereotactic radio-surgery and having

greater morbidity, brachytherapy in malignant gliomas can be used for

infiltrating or cavitary tumors and for tumors larger than 3 cm.

Brachytherapy can be done as a boost to external beam RT or as salvage

therapy at recurrence. It was shown to improve survival in patients with

malignant gliomas.[89,90] Serious complications after brachytherapy are

wound infections, cerebral edema, abscess into the tumor, hemorrhage, and

radiation necrosis that requires surgical intervention. These complications

make brachytherapy a less attractive treatment modality for elderly patients

and for patients with poor performance status.

Radiation beams other than the usual photons and electrons are now under

investigation. Beams of protons, neutrons, and Pi-mesons provide better dose

localization on the tumor as well as radiobiological efficiency, while

sparing the surrounding normal tissues. Radioimmunotherapy is another method

to ensure dose localization, in which a monoclonal antibody coupled with a

radionuclide is introduced into the tumor. [91] The monoclonal antibody is

designed to bind to receptors expressed only by tumor and not by normal

cells, thus sparing normal tissue.

Boron neutron capture therapy (BNCT) is a form of RT presently under

investigation for treatment of malignant gliomas. [92] BNCT is mediated by

short-range (less than 10 microns), high-energy particles resulting from

neutron-induced disintegration of boron-10. There is preferential

accumulation of boron-10 in conjunction with high thermal neutron flux at

the tumor site. The bombardment of the boron nucleus with a slow neutron

induces disintegration of boron, which yields ionizing radiation. Best

results with BNCT were reported by Japanese investigators. [93] The studies

do not specify differences, if any, in response based on tumor types, age,

and performance status. The initial clinical trials have been marred by

significant brain necrosis. The improved technologies have rekindled the

interest in this treatment modality. [92]

Side Effects of Radiation Therapy

Regardless of the radiation modality used, RT may cause side effects that

need to be discussed with patients and monitored. The reactions to RT are

more significant when RT is administered to a large portion of the brain.

The effects can be acute, early delayed, and late delayed. [94] Acute

effects occur during treatment or shortly after completion of RT. Some

patients experience headaches, probably related to edema, or a worsening of

the neurologic deficits. Fatigue is another complaint and, depending on the

tumor location, patients may experience nausea, sore throat, hearing loss,

or blurred vision. These symptoms are transient and can be controlled with

corticosteroids and reassurance. Early-delayed effects, which appear in the

first 3 months after completion of RT, include somnolence, loss of appetite,

and apathy. These effects are self-limiting and seem to be more severe in

older patients. Late-delayed radiation injury occurs months or even years

after completion of radiation. Patients experience short-term memory loss

and cognitive decline. [94-99] CT or MRI scans reveal white matter changes

bilaterally or may show focal radiation necrosis. [95] Areas of necrosis do

enhance and can have surrounding edema, which is difficult to distinguish

from recurrent tumor. [96] PET and SPECT scans can be helpful, showing

metabolically hypoactive areas of radiation necrosis compared with the

increased metabolic activity in tumor tissue.[37,38] The definitive

differential diagnosis is done by biopsy. [97] The degree of cognitive

impairment can be more severe in patients with tumors in the temporal lobes

and in elderly patients with baseline mild dementia. In the latter patient

population, the decision regarding RT must be based on tumor type and life

expectancy. The degree of cognitive impairment can be quantitated using

neuropsychometric evaluation prior to RT and at intervals of 6, 12, and 18

months. [97,99]

Chemotherapy

Chemotherapy is now established as accepted treatment for primary brain

tumors. [100-102] The addition of chemotherapy to RT has been shown to

prolong survival by another 6-18 months, depending on the grade of the

tumor. Longer-term survivals have also been reported. While multiple

clinical trials are investigating new drugs and drug combinations, only a

small proportion of the overall population with brain tumors, and in

particular patients over 65 years of age, are entered in clinical trials.

Several factors determine chemotherapy failure. Brain tumors are

heterogeneous, and glioma cells have been shown to express the multidrug

resistance gene (MDR-1). [103] Repair enzymes such as

glutathione-S-transferase and O 6 -alkylguanine alkyl transferase counteract

the cytotoxic effect of platinum compounds and nitrosoureas; some of the

tumor cells are in the G0 phase and are less susceptible to chemotherapy.

[104,105] The DNA mismatch repair deficiency has been identified as an

important mechanism conferring resistance to RT and methylating agents such

as procarbazine and temozolomide. [105] The blood-brain barrier limits the

brain tissue penetrance of drugs that are nonionized or are not liposoluble.

The drugs should attain and maintain a cytocidal concentration in the tumor.

[106-108] This is dependent on the physical and chemical properties of the

drug and its pharmacokinetics. The half-life of the drug, intracellular

binding, and capillary-to-cell diffusion are important factors. The dose,

route of administration, and schedule of administration can influence the

drug concentrations.

Several classes of drugs are used for the chemotherapy of brain tumors:

alkylating agents, antimetabolites, natural compounds, urea analogs,

methylhydrazine derivatives, and polyamine inhibitors. [102] Nitrosoureas

are to date the most effective drugs for treatment of malignant gliomas.

[109-117] The two most widely used nitrosoureas are

1,3bis-(2-chloroethyl)-1-nitrosourea (BCNU; carmustine), which is

administered intravenously or intraarterially, and

1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea (CCNU; lomustine), given

orally. The clinical activity is similar for the two drugs, with 40%

response rates. Resistance to BCNU is determined by the activity of the

enzyme O 6 -alkylguanyl alkyl transferase, which is not age dependent. [104]

The dose-limiting toxicities are myelotoxicity and pulmonary fibrosis.

Patients with chronic obstructive lung disease must be monitored closely

during the treatment. Platinum compounds (cisplatin and

carboplatin),[109,110] procarbazine, [111-115] and etoposide are also

effective but do not offer a significant survival advantage over BCNU.

Currently, chemotherapy with combined procarbazine, CCNU, and vincristine

(PCV) is considered to be the most effective for low-grade or anaplastic

oligodendrogliomas and for anaplastic astrocytomas. [113,114] Recent

analysis, however, shows no clear survival advantage with PCV over BCNU.

[115] In low-grade oligodendrogliomas, chemotherapy with PCV was shown to

inhibit tumor growth and induce regression of the tumor on imaging studies.

[116]

Temozolomide (Temodal), which has been recently approved by the Food and

Drug Administration for the treatment of recurrent anaplastic gliomas, is

still undergoing clinical trials in combination with RT and other

chemotherapeutic agents. It is administered orally and is well tolerated

with no significant myelotoxicity. It can be safely administered to elderly

patients, and the efficacy does not appear to be age related. [117]

Chemotherapy is used traditionally after completion of RT as adjuvant

treatment or at the time of tumor recurrence or progression. [118-120] Some

chemotherapeutic agents (eg, BCNU, hydroxyurea, and vincristine) also have a

radiosensitizing effect, and there are clinical trials using chemotherapy in

conjunction with RT. [121] Recent reports suggest a benefit in using

chemotherapy prior to RT, particularly in oligodendrogliomas. [122] Newer

agents such as the topoisomerase I inhibitors topotecan and irinotecan

(CPT-11) are also under investigation as preradiation chemotherapy for

glioblastomas. Preradiation chemotherapy can be used for palliation in

patients with low performance status. [122]

The most common side effect of chemotherapy is myelosuppression, which may

require blood transfusions or the use of colony-stimulating factors.

Myelo-suppression from chemotherapy occurs earlier in the course of

treatment in older patients. No significant nitrosourea-induced pulmonary

toxicity was noted in patients over the age of 60, possibly because their

survival rates are low, and pulmonary fibrosis occurs after several courses

of treatment. Other common side effects of chemotherapy are nausea, fatigue,

and loss of appetite. These side effects are mild, are usually

self-limiting, and respond well to symptomatic treatment. Procarbazine can

cause allergic reactions or, if dietary restrictions are not observed, can

cause paroxysmal hypertension. Peripheral neuropathy is a common side effect

of vincristine, cisplatin, and procarbazine. The symptoms are numbness in

the hands and feet, constipation, and occasionally jaw pain or numbness.

When the neuropathy affects the fine motor skills, the offending agent must

be discontinued. Preexisting conditions such as diabetes, hypothyroidism,

and vitamin B12 deficiency cause peripheral neuropathy. To avoid

debilitating neuropathy, patients need to be evaluated neurologically before

starting chemotherapy with these drugs. Patients treated with etoposide are

at higher risk for second malignancies, mainly leukemias. In patients with

malignant gliomas, long-term survival is low and does not allow enough time

for a second malignancy to develop. These side effects are noted in all age

groups.

Hormonal Therapy

Laboratory studies have shown that protein kinase C (PKC) is an important

factor in promoting proliferation of malignant gliomas. Tamoxifen, an

estrogen-receptor blocking agent commonly used in treating breast cancer,

has been shown to inhibit proliferation of malignant astrocytomas via

nonestrogen-receptor-mediated PKC blockade. In clinical studies, the dose of

tamoxifen shown to inhibit brain tumor growth is much higher than the dose

used for breast cancer (40-100 mg b.i.d. vs 10 mg b.i.d.). The effect

appears to be dose dependent and is cytostatic rather than cytotoxic. Still,

in patients with good performance status, tamoxifen has been shown to

increase survival. The antimitotic effect is not reversed by estrogen,

indicating a nonestrogen-receptor-mediated mechanism of action. Tamoxifen

does cross the blood-brain barrier[123,124] and is well tolerated, even at

these high doses. Tamoxifen is now being studied in combination with BCNU as

adjuvant therapy after RT. Because of its good safety profile and ease of

administration, tamoxifen can be offered as an alternative treatment to

elderly patients with malignant gliomas who had received RT and do not wish

to take chemotherapy but would consider other forms of treatment. The

incidence of thromboembolic complication from tamoxifen is higher in the

brain tumor patients than in breast cancer patients, but these patients have

an overall higher incidence of thromboembolism, even without tamoxifen.

Biologic Therapy

Biologic therapies with differentiating and immunomodulatory agents are

presently under investigation as alternatives to the conventional forms of

treatment.[125,126] They can be used alone or in combination with RT or

chemotherapy. These agents are cytostatic rather than cytotoxic. The

mechanisms of action are not yet completely elucidated.

Retinoids. The retinoids -- 13-cis-retinoic acid (CRA) and

all-trans-retinoic acid (TRA) -- are natural and synthetic derivatives of

vitamin A and have proven efficacy in some premalignant and malignant

conditions. [127] In vitro studies demonstrated the differentiating and

growth inhibitory effect of retinoic acid on glioma cells. The growth

inhibition is related to a decrease in epidermal growth factor

receptor-mediated phosphorylation activity. In clinical studies, CRA showed

activity against malignant gliomas.[128,129] The side effects were

relatively mild: dryness of the skin and mucosa and headache. In some

patients, headaches appeared to be due to increased intracranial pressure

(pseudotumor cerebri-type) and responded to treatment with diuretics and

glycerol. Another complication of treatment with retinoids that is

potentially fatal is pancreatitis, and the enzymes amylase and lipase must

be monitored carefully during therapy. Both TRA and CRA are administered

orally in doses of 60-120 mg/m [2] per day for 3 weeks, followed by 1 week

of rest.

Immunomodulators. Interferons (IFNs) are naturally produced glycoproteins

with antiviral, antiproliferative, and immunomodulatory properties. [130]

Both IFN-alpha and IFN-beta have demonstrated activity against malignant

gliomas in vitro and in clinical trials. [130-133] IFN-alpha-2b modulates

the activity of PKC by downregulating it. [130] There are reports of

enhanced activity of IFN and other agents in combination against various

tumors, as well as an antiangiogenic effect. [133-135]

Known side effects of IFN therapy are flu-like symptoms and hypotension.

Some patients develop low back pain or arthralgia that at times can be

severe enough to warrant discontinuing the treatment. This can make IFN less

well tolerated by elderly patients with arthritis.

------------------------------------------------------------------------

New Treatments for Malignant Gliomas

The management of brain tumors continues to bring new challenges for the

treating physicians, and ongoing research is aimed at defining the biologic

mechanisms of malignancy and developing new treatment modalities. Managed

care policies and the need for cost containment make treatment decisions

even more difficult, especially for elderly patients.

Modification of Conventional Therapies

While there has been significant progress in identifying the genes

responsible for oncogenesis and drug resistance, finding effective

treatments, particularly for malignant gliomas, has been less successful.

Some studies combine conventional therapies with novel approaches, while

others introduce new experimental treatments.

Factors that influence the efficacy of chemotherapeutic agents are the

unique cytoarchitecture of the brain (with an effective blood-brain

barrier), the infiltrative pattern of growth of some primary brain tumors,

and the heterogeneity of tumor cells. New approaches to chemotherapy are

directed at circumventing the blood-brain barrier, overcoming drug

resistance mechanisms, and minimizing the systemic and neurotoxicity.

The blood-brain barrier can be circumvented by increasing the dose of the

drug (with subsequent increased toxicity), by administering intraarterial

chemotherapy with blood-brain barrier modification, or by using new delivery

systems. Intratumoral administration has the advantage of allowing delivery

of cytotoxic concentrations of drugs in the tumor bed, with no systemic

toxicity. The disadvantages are related to diffusion problems and local

toxicity (necrosis). RMP-7 is a bradykinin analog that opens the blood-brain

barrier selectively and increases penetration of chemotherapeutic agents

into the brain. [136-137] Chemotherapy can be administered intratumorally

via an Ommaya reservoir or through liposomes or biodegradable polymers. BCNU

wafers (Gliadel) are now available commercially and can be placed into the

tumor bed at the time of resection. [138]

Novel Therapies

Gene therapy refers to the introduction of new genetic material into cells

to provide beneficial effect to the patient. The preferred method for gene

transfer is through viral vectors, which have a high efficiency in infecting

host cells by inserting their own genetic material into the host cell

genome. Both retroviruses and adenoviruses are studied for use as vectors

for gene therapy. [139,140]

The most publicized gene therapy clinical trial for brain tumors involves

transfer of the herpes simplex virus thymidine kinase (HStk) gene into tumor

cells, using retroviral vectors. HStk is an enzyme that phosphorylates the

antiviral prodrug gancyclovir, which becomes virucidal and cytotoxic. [140]

Few animal and clinical data are available regarding the efficacy of gene

therapy for brain tumors, the long-term effects, and the safety of viral

vectors.

Immunoconjugates are cytotoxic compounds that combine a ligand and a

cytotoxic agent, which can be a radioisotope, a chemotherapeutic drug, or a

toxin. [141] Clinical trials have been conducted on small numbers of

patients with leptomeningeal carcinomatosis using immunoradiotherapy. [142]

Current clinical trials are underway for treatment of malignant gliomas.

Glioma cells express transferrin receptors, which are targeted with

immunoconjugates using as ligands either monoclonal antibodies to

transferrin receptors, or transfer-rin, conjugated with a toxin. The toxins

are ribosomal inhibitors, either of plant (ricin) or bacterial (CRM-107)

origin. The dose of toxin necessary for tumoricidal effect is smaller than

predicted by the tumor volume, which indicates a bystander effect. The

results of these trials are not yet available.[141,142]

Recent studies evaluate other biologic agents for treatment of malignant

gliomas, targeting factors that intervene in cell replication or

angiogenesis.

Malignant gliomas utilize mevalonate for synthesis of cholesterol and

intermediates for cell replication. Lovastatin and phenylacetates inhibit

the enzymes HMG-CoA reductase and MVA-PP decarboxylase and thus affect the

mevalonate synthesis and utilization and induce cytostasis and apoptosis.

Both lovastatin and phenylacetates are presently in clinical

trials.[143,144]

Angiogenesis is an important feature in malignant gliomas. There are

multiple angiogenesis factors that can be targeted specifically. Fumagillin,

an antibiotic derived from the fungus Aspergillus fumigatus Fresenius, was

noted to inhibit angiogenesis in vitro. [145] Endostatin and AGM-1470,

synthetic analogs of fumagillin that are more potent and less toxic, are now

in early clinical trials, and their efficacy is yet to be proven. [146]

Thalidomide is an antiangiogenic drug that, in combination with carboplatin,

has shown activity against anaplastic gliomas. Thalidomide is also now in

clinical trials for meningiomas. Its mild side effects profile makes it an

attractive drug for treatment of tumors in elderly patients.

These novel therapies are still in the experimental phase. Larger studies

and longer follow-up periods will be necessary to evaluate the safety and

efficacy of these new therapies against brain tumors in all age groups as

well as the differences, if any, in older patients.

Treatment of Primary Central Nervous System Lymphoma

In nonimmunocompromised patients, primary central nervous system lymphoma is

a disease of the elder-ly. The management combines systemic and intrathecal

chemotherapy with RT. [147] Clinical studies show that elderly patients

tolerate intensive chemotherapy well. However, prognosis remains poor, with

an average survival of only 1 year.

Treatment of Brain Metastases

The treatment modalities are similar for primary and metastatic brain

tumors. For brain metastases, the choice of treatment is based also on the

status of the systemic disease. When feasible, surgical resection will

significantly improve the performance status and prolong survival. [148-150]

Brain metastases are optimal lesions for radiosurgery. Chemotherapy can be

considered for control of both brain and active systemic metastatic disease.

[148]

Link to comment
Share on other sites

Join the conversation

You are posting as a guest. If you have an account, sign in now to post with your account.
Note: Your post will require moderator approval before it will be visible.

Guest
Reply to this topic...

×   Pasted as rich text.   Paste as plain text instead

  Only 75 emoji are allowed.

×   Your link has been automatically embedded.   Display as a link instead

×   Your previous content has been restored.   Clear editor

×   You cannot paste images directly. Upload or insert images from URL.

Loading...
×
×
  • Create New...