Brain Metastases and Neoplastic Meningitis


Summary of Key Points

  • Incidence

  • Central nervous system (CNS) metastases are common, affecting as many as 20% of patients with cancer.

  • Less often, the dura, leptomeninges, skull base, or cranial nerves may be affected.

  • The most frequent primary tumor types that give rise to brain metastases include lung cancer, melanoma, breast cancer, and renal cell carcinoma.

  • Leptomeningeal dissemination (neoplastic meningitis) can occur in patients without known brain parenchymal metastases.

  • Neoplastic meningitis is mostly seen in patients with breast cancer, lung cancer, and melanoma.

  • Diagnosis

  • Contrast-enhanced magnetic resonance imaging (MRI) is the best method to diagnose malignant CNS involvement.

  • Metastases generally appear as enhancing, well-circumscribed lesions with or without surrounding vasogenic edema. In patients with neoplastic meningitis, meningeal enhancement is only visible on MRI in about 50% of cases.

  • Biopsy or resection may be indicated to confirm the diagnosis of metastases.

  • Cerebrospinal fluid (CSF) cytology in cases of neoplastic meningitis may be negative initially in 40% to 50% of cases.

  • Contrast-enhanced MRI should be performed before spinal puncture for CSF evaluation because it can result in transient leptomeningeal enhancement on MRI.

  • Prognosis

  • Survival of patients with brain metastases has a wide range from days to years.

  • Prognostic indices such as the Graded Prognostic Assessment (GPA) can assist in estimating survival and assist in appropriate treatment decisions.

  • Neoplastic meningitis tends to have grave prognosis with a median survival of 3 months.

  • Poor performance status is associated with poor prognosis in patients with brain metastases and those with leptomeningeal involvement.

  • Treatment

  • A multidisciplinary approach should be applied for patients with any type of CNS involvement.

  • Patients with a good prognosis and a limited number of brain metastases may benefit from more aggressive therapy such as surgery or stereotactic radiation therapy (SRT), with or without adjuvant whole-brain radiotherapy (WBRT).

  • Cognitive decline after WBRT has led to omission of WBRT in selected patients with brain metastases and to greater use of SRT.

  • Treatment of leptomeningeal disease mainly includes intrathecal chemotherapy, radiation, and systemic chemotherapy.

  • The role of targeted treatments for brain metastases and leptomeningeal disease is evolving.

Central nervous system (CNS) metastases result from the intracranial spread of tumor cells that originate outside of the CNS. Most CNS metastases involve the brain parenchyma, dura, or leptomeninges; less commonly, the skull base, cranial nerves, or dural sinuses are involved.

Metastases to the CNS can cause substantial morbidity and mortality. Currently, there are many exciting changes with regards to our understanding of tumor genomics, molecular subtypes, immunogenicity, and advances in systemic treatments and radiation therapy (RT) technology and delivery. Prognostic tools have evolved, with the understanding that unique features of the primary tumor have significance for the prognosis of patients with brain metastases. Brain metastases are no longer a single entity but are assessed and managed in the context of the primary tumor. Moreover, advances in systemic treatments can prolong extracranial disease control, potentially identifying patients who might benefit from a more aggressive treatment for brain metastases. Some of the new systemic treatments are thought to contribute to intracranial control or intracranial complications after RT. Advances in technology and RT delivery allow delivering high doses with significant precision and accuracy (i.e., stereotactic radiation therapy [SRT]), thus minimizing the radiation doses to normal tissues. In our view, the future of RT for CNS metastases (parenchymal and leptomeningeal metastases) will change significantly. We provide a review of existing evidence-based literature that is the basis for current treatment guidelines and discuss potential future developments.

Brain Metastases

Epidemiology

Brain metastases are estimated to be 10 times more common than primary malignant brain tumors in adults, occurring in 6% to 20% of patients with cancer. However, the exact incidence is unknown because the incidence is derived from disparate data sources (e.g., cancer registries, death certificates) and different type of studies (e.g., population, clinical, autopsy based). Nevertheless, the reported incidence of brain metastases is increasing probably because of improvement of neuroimaging, increased surveillance, and improved control of systemic disease, resulting in prolonged survival.

The most common primary tumors responsible for brain metastases include lung cancer (30% to 80%), breast cancer (5% to 30%), melanoma (5% to 21%), renal cell carcinoma (5.5% to 11%), and gastrointestinal tract cancer (e.g., colorectal cancer, 1.4% to 4.8%). Although brain metastases may arise from any primary cancer, some tumors, such as prostate, oropharyngeal, and skin carcinomas, rarely metastasize to the brain.

Cancer may spread to the brain at various points in the course of disease, but the timeline differs among tumor types. Symptomatic brain metastases are at times the presenting symptoms that lead to the diagnosis of lung cancer. In other cases, the median interval between diagnosis of lung cancer and identification of brain metastases ranges from 2 to 9 months, with about 90% of the lung cancer brain metastases diagnosed within 1 year of initial diagnosis of lung cancer. But in breast cancer, 80% of the patients with CNS involvement are diagnosed after systemic spread was diagnosed, occurring at an interval of 1 to 3 years after initial diagnosis.

As previously stated, one of the reasons the incidence of brain metastases is increasing is attributed to success in controlling systemic disease and prolonged survival. A prime example was seen after the introduction of trastuzumab (anti–human epidermal growth factor receptor 2 [HER2]), which does not cross the blood–brain barrier, the incidence of brain metastases in HER2-positive breast cancer patients increased from 1.6% to 10.5%, mostly because of improved survival provided by trastuzumab.

Other factors suggested to contribute to the incidence of brain metastases include advanced disease stage, gender, race, and tumor-related factors. Within a primary tumor type (e.g., lung, breast, and melanoma), factors such as histology, tumor grade, molecular subtype, and mutation status influence the incidence of brain metastases. For example, most lung cancer brain metastases are secondary to small cell lung cancer (SCLC) and adenocarcinoma ; breast cancer types with higher probability to metastasize to the brain include high-grade cancers, HER2-positive (irrespective of systemic treatment), and triple-negative subtypes. Melanoma subtypes associated with increased risk for brain metastases include mucosal melanoma, head and neck or trunk primary cutaneous melanomas, acral lentiginous or nodular melanoma histologies, and melanoma patients with BRAF or NRAS mutations.

Prevention and Early Detection

It is not clear if screening for brain metastases is beneficial in asymptomatic solid tumor patients; however, because of the high incidence of brain metastases in non–small cell lung cancer (NSCLC) and SCLC, current guidelines recommend brain imaging (magnetic resonance imaging [MRI] with contrast is preferred) at an initial diagnosis of SCLC and for patients with NSCLC stage II and more (optional for stage IB). The decision to perform neuroimaging in patients with other cancer type without known brain metastases should be based on clinical suspicion.

Similarly, because of the high incidence of brain metastases early after initial cancer diagnosis, elective brain irradiation (PCI [prophylactic cranial irradiation]) was suggested for patients with lung cancer in an attempt to reduce the incidence of overt brain metastases, prolong survival, and improve quality of life. This approach was proposed to overcome the brain as a sanctuary site because of the poor penetrance of systemic drugs to the brain parenchyma. The concept of PCI was also evaluated for other solid tumors (e.g., breast cancer) but without clear therapeutic benefit.

There is some evidence that targeted therapies can prevent, delay, or treat early CNS metastases. A phase III trial evaluating capecitabine with or without lapatinib in advanced HER2-positive breast cancer showed that patients treated with capecitabine and lapatinib had a lower rate of CNS disease compared with lapatinib alone (4% versus 13%; P = .45). Whether better systemic control by capecitabine and lapatinib resulted in a lower rate of CNS involvement is unclear. Currently, there are a few ongoing trials evaluating systemic therapies (mainly targeted agents) in reducing the incidence of brain metastases.

Pathophysiology

Brain metastases arise primarily from arterial hematogenous spread. Large aggregates of tumor cells that gain access to the venous circulation are filtered out in lung capillaries before entering the systemic arterial circulation, but individual tumor cells may pass through and lodge in the brain. Tumor cells tend to become trapped where blood vessels decrease in caliber at the gray–white matter junction and distalmost vasculature (the border or “watershed” zones). Metastatic cells then adhere to the endothelial cells, penetrate into the brain parenchyma, and proliferate.

It has been suggested the tumor emboli may also break off from lung metastases or primary lung cancers to travel to the brain via the arterial circulation. Cells from pelvic or abdominal cancers may gain access to the posterior fossa or leptomeninges through Batson's vertebral venous plexus without passing through the lungs.

The distribution of metastases is roughly proportional to the relative blood flow to different regions; approximately 80% of brain metastases are located in the cerebral hemispheres, 10% to 15% in the cerebellum, and 1% to 5% in the brainstem. Posterior fossa metastases appear to arise disproportionately from pelvic or abdominal primary tumors.

Most brain metastases are very well circumscribed. Although extensive associated edema may be present, the tumor cells do not tend to infiltrate into surrounding brain tissue, in contrast to primary malignant brain tumors. Most brain metastases are solid, but they may be heterogeneously enhancing or cystic because of necrosis, keratin deposits in squamous cell carcinoma, or mucin secretion from adenocarcinomas. Brain metastases may be hemorrhagic, particularly from melanoma, renal cell carcinoma, choriocarcinoma, and, less frequently, bronchogenic carcinoma.

Clinical Presentation

The possibility of brain metastases should be suspected in any patient with cancer who experiences new neurologic signs or symptoms. Symptoms often worsen gradually over time from a growing tumor and associated edema. Less often, acute neurologic symptoms may occur as a result of hemorrhage. The most common presenting symptoms are fatigue, headache (24% to 53%), focal weakness (16% to 40%), altered mental status (24% to 31%), gait ataxia (9% to 21%), seizures (15% to 18%), and difficulty in speech (12%) depending on the location of the lesion within the brain parenchyma and degree of edema. Other common symptoms include insomnia, mood changes, and difficulty in concentrating, loss of appetite, and nausea. Patients who present with drowsiness or dyspnea tend to have a poor survival and might need urgent intervention (e.g., corticosteroids). Symptoms may worsen after treatment, and potentially reversible processes such as increasing edema from treatment should be suspected and treated accordingly. Severe symptoms and worsening of symptoms can serve as a prognostic tool and assist the physician to set appropriate care goals with the patient and her or his caregiver, plan an appropriate treatment approach, and have time to address end-of-life concerns.

Diagnosis

It is widely accepted that MRI is the best diagnostic to detect brain metastases and evaluate the number, size, and location of metastases. Standard MRI includes T2-weighted and pre- and postgadolinium-enhanced T1-weighted sequences. A postcontrast fluid-attenuated inversion-recovery (FLAIR) sequence is helpful in visualizing small metastases near cerebrospinal fluid (CSF) spaces and evaluate the extent of edema ( Fig. 55.1 ). High-dose gadolinium and 1-mm slices may aid in visualizing small brain metastases and are also useful for SRT treatment planning. The role of MRI in characterizing brain metastases is evolving. Functional imaging techniques, such as magnetic resonance spectroscopy and perfusion and diffusion, may aid in distinguishing metastatic lesions from other enhancing lesions such as a primary brain tumor or nonmalignant processes. New studies suggest the potential use of morphology and texture analysis of MRI in differentiating between histologic subtype within a given group (e.g., lung adenocarcinoma versus squamous cell, subtypes of breast cancer).

Figure 55.1, Typical appearance of a brain metastasis on magnetic resonance imaging. The lesion is well-circumscribed and brightly enhancing on the postcontrast T1-weighted image (left) . Both edema and cerebrospinal fluid (CSF) show up as increased signal on the T2-weighted image (center) . Both the metastasis and surrounding edema appear bright on the postcontrast fluid-attenuated inversion-recovery image, but the CSF signal is suppressed (right) .

In patients who were previously treated for brain metastasis or exposed to RT, the differential diagnosis of an enhancing brain lesion also includes radiation necrosis. The location of the lesion with respect to previous radiation treatment can aid in the diagnosis; however, it is often difficult to differentiate between the two, and additional diagnostic evaluation may be needed. These may include MRI spectroscopy, perfusion imaging, or both. Positron emission tomography (PET) using fluorodeoxyglucose (18F or FDG) has low sensitivity and specificity in the brain but may help in differentiating viable tumor from radiation necrosis. Correlation or coregistration with contrast-enhanced-MRI can increase the diagnostic accuracy of PET-FDG in these cases. Other amino acid–based PETs (e.g., O-(2-[18F]fluoroethyl)-L-tyrosine [FET], L-[methyl-11C]methionine [MET]) were also shown to be beneficial in diagnosing brain lesions and differentiating viable tumor from necrosis. Delayed contrast and perfusion MRI (TRAMs) is a new method that can aid in differentiating necrosis from viable tumor and was shown to correlate with histologic findings of viable tumor or necrosis in patients with primary and metastatic brain lesions.

The differential diagnosis of an enhancing or hemorrhagic intracranial lesion also includes primary brain tumor, CNS lymphoma, abscess, encephalitis, cerebral infarct or hemorrhage, progressive multifocal leukoencephalopathy, multiple sclerosis, and demyelinating disease. However, tissue confirmation (biopsy, resection) may be warranted if there is doubt about the diagnosis or if a solitary brain lesion seen in a patient without a history of cancer (or with other accessible lesions) or a history of cancer but no other known metastatic disease.

Even if the diagnosis seems obvious in patients with a known cancer, tissue confirmation might have significant implications. In Patchell's 1990 publication that established the role of surgery before whole-brain radiotherapy (WBRT) in patients with a known systemic cancer, 11% of 54 patients with a single brain lesion that was thought to be a metastasis turned out to have glioblastoma, low-grade astrocytoma, abscess, or an inflammatory process. Imaging has much improved to aid in diagnosis, but the differential diagnosis of an enhancing brain lesion should be kept in mind.

Prognostic Factors

In general, brain metastases are associated with a poor prognosis. Despite major advances in cancer diagnosis, cancer treatment, and brain imaging, the overall survival time of patients with brain metastases treated with WBRT without surgery or stereotactic radiosurgery (single fraction, SRS) has remained at about 2 to 6 months since the 1950s. Although some patients will survive 1 to 2 years, only approximately 2.5% will survive more than 5 years with aggressive treatments.

Prognostic factors have been evaluated in several large series. Gaspar and colleagues identified three prognostic groups by using a recursive partitioning analysis (RPA) of more than 1100 patients enrolled in three consecutive Radiation Therapy Oncology Group (RTOG) trials. The median survival period was 7.1 months for RPA class 1, consisting of patients younger than 65 years old with a Karnofsky performance status (KPS) of 70 or greater, controlled primary tumor, and no extracranial metastases. Patients with KPS less than 70 (RPA class 3) had a median survival period of only 2.3 months; the median survival period was 4.2 months for the remaining patients (RPA class 2). Yamamoto and colleagues suggested a subclassification of the RPA II into three categories to better identify patients who might benefit from more aggressive approach.

Sperduto and colleagues developed a newer prognostic index for patients with brain metastases termed the Graded Prognostic Assessment (GPA) based on a database of 1960 patients accrued to several RTOG brain metastasis protocols that was validated and refined with diagnosis-specific (DS-GPA) prognostic indices based on an independent, multi-institutional retrospective analysis of 4259 other patients with brain metastases treated with WBRT, SRS, or both ( Tables 55.1 and 55.2 ). Sperduto and colleagues demonstrated a wide range of survival (from 3.4 months to 25.3 months) for breast cancer patients with brain metastases. The prognostic analysis for breast cancer patients was revised to include primary tumor subtype that was significant for survival.

Table 55.1
Disease-Specific Graded Prognosis Analysis
Worksheet modified from Sperduto, et al. Summary report on the graded prognostic assessment: an accurate and facile diagnosis-specific tool to estimate survival for patients with brain metastases. J Clin Oncol 2012;30(4):419–25.
N on –S mall C ell and S mall C ell L ung C ancer
Prognostic Factor Scoring Criteria Patient Score
0 0.5 1.0
Age (y) >60 50–60 <50
KPS <70 70–80 90–100
Extracranial metastases Present Absent
Number of brain metastases >3 2–3 1
Total:
Median survival (months) by score: 0–1.0 = 3.0; 1.5–2.0 = 5.5; 2.5–3.0 = 9.4; 3.5–4.0 = 14.8
Prognostic Factor Scoring Criteria Patient Score
0 1.0 2.0
KPS <70 70–80 90–100
Number of brain metastases >3 2–3 1
Total:
Median survival (months) by score: 0–1.0 = 3.4; 1.5–2.0 = 4.7; 2.5–3.0 = 8.8; 3.5–4.0 = 13.2
Prognostic Factor Scoring Criteria Patient Score
0 1.0 2.0
KPS <70 70–80 90–100
Number of brain metastases >3 2–3 1
Total:
Median survival (months) by score: 0–1.0 = 3.3; 1.5–2.0 = 7.3; 2.5–3.0 = 11.3; 3.5–4.0 = 14.8
B reast C ancer

Prognostic Factor Scoring Criteria Patient Score
0 0.5 1.0 1.5 2.0
KPS ≤50 60 70–80 90–100 N/A
Subtype a Basal N/A Luminal A HER2 Luminal B
Age (y) ≥60 <60 N/A N/A N/A
Total:
Median survival (months) by score: 0–1.0 = 3.4; 1.5–2.0 = 7.7; 2.5–3.0 = 15.1; 3.5–4.0 = 25.3
G astrointestinal C ancers
Prognostic Factor Scoring Criteria Patients Score
0 1.0 2.0 3.0 4.0
KPS <70 70 80 90 100
Total:
Median survival (months) by score: 0–1.0 = 3.1; 2.0 = 4.4; 3.0 = 6.9; 4.0 = 13.5

ER, Estrogen receptor; HER2, human epidermal growth factor receptor 2; KPS, Karnofsky performance score.

a Subtype: Basal: triple negative; luminal A: ER/PR positive, HER2 negative; luminal B: triple positive; HER2: ER/PR negative, HER2 positive.

Table 55.2
Median Survival Time a for Patients With Brain Metastases by DS-GPA
Modified from Sperduto PW, Kased N, Roberge D, et al. Summary report on the graded prognostic assessment: an accurate and facile diagnosis-specific tool to estimate survival for patients with brain metastases. J Clin Oncol. 2012;30(4):419–425.
Primary Tumor Median Survival (mo) GPA S core
0–1 1.5–2.0 2.5–3.0 3.5–4.0
NSCLC 7 3 5.49 9.43 14.78
SCLC 4.9 2.79 4.90 7.67 17.05
Melanoma 6.74 3.38 4.7 8.77 13.23
RCC 9.63 3.27 7.29 11.27 14.77
Breast cancer 13.8 3.35 7.7 15.07 25.3
GI cancer 5.36 3.13 4.4 6.87 13.54
DS-GPA, Disease specific Graded Prognostic Assessment; GI, gastrointestinal; N/A, not applicable; NSCLC, non–small cell lung cancer; RCC, renal cell carcinoma; SCLC, small cell lung cancer.

a Survival before use of targeted treatments (e.g., immunotherapy). Confidence interval available in original publication.

Treatment

Corticosteroids

Glucocorticoid therapy is generally instituted in symptomatic patients as soon as brain metastases are diagnosed to help alleviate symptoms from cerebral edema. Glucocorticoids reduce the permeability of leaky tumor blood vessels and thereby reduce the mass effect and edema caused by brain metastases. The most commonly used steroid is dexamethasone because of its relatively low mineralocorticoid activity and long half-time (~36–54 hours); after oral use, it is readily absorbed. A randomized trial suggested that a 4- or 8-mg dose of dexamethasone is as effective as 16 mg. Therefore 4 or 8 mg/day of dexamethasone divided into two doses is suitable as an initial dose. Higher doses are recommended in patients with severe mass effects symptoms or who do not respond to treatment within 48 hours. After patients become asymptomatic or reach maximal benefit, the dose should be gradually tapered and either discontinued or maintained at the lowest dose level needed to manage symptoms. Abrupt withdrawal after prolong use should be avoided. If headaches recur or neurologic symptoms worsen during the course of the taper, the dose should be increased as needed and then the taper should proceed more gradually.

If CNS lymphoma is suspected via imaging, is it is advised to hold steroids until tissue diagnosis is available. CNS lymphoma is extremely sensitive to corticosteroids, resulting in tumor cell lysis and regression of tumor. The speed of regression is variable, but a complete disappearance may occur in 1 to 2 days.

Anticonvulsant Agents

About 15% of patients with brain metastases present with seizures when they are first seen by a clinician, and 30% to 40% experience seizures at some point in their disease course. An anticonvulsant agent should be prescribed for any patient with brain metastases who experiences a seizure. Prophylactic anticonvulsant agents are not recommended; prospective and retrospective studies and meta-analyses have failed to demonstrate a benefit for prophylactic anticonvulsant agents in patients with brain metastases. Short-term prophylactic anticonvulsants treatment can be used in patients who are undergoing craniotomy and resection. It is recommended to start tapering and discontinuing the anticonvulsants after the first postoperative week, especially if the patient is medically stable. Anticonvulsant agents can have adverse side effects, and some may reduce the efficacy of corticosteroids and can activate the cytochrome P450 enzyme system. This latter property can affect patients undergoing chemotherapy by altering the metabolism of some chemotherapeutic agents, thus requiring chemotherapy dose adjustment.

Definitive Treatment

Treatment of brain metastases is selected based on the number, size (and total volume of intracranial disease), and location of the brain lesions. Patients' symptoms, performance status, extent and control of systemic disease have prognostic implications that should be considered in planning treatment. Prognostic tools such as the DS-GPA are useful to help the clinician set appropriate care goals.

Surgery

Surgery can play a pivotal role in the management of brain metastases. At the time of presentation, a neurosurgeon should be consulted. Surgery may be needed for tissue confirmation, relieving mass effect from a large symptomatic lesion, improving the likelihood of durable local control for a single metastasis (especially metastasis >3 cm), and salvaging a metastasis that failed to respond to prior therapy. Surgery can also assist in cases of large cystic lesions that are planned for SRT by allowing rapid decompression of a cyst or clot, leading to reduction of swelling and reducing the target volume.

Resection of brain metastases has become safer with advances in neuroimaging and neurosurgery such as image guidance, preoperative and intraoperative functional mapping, and intraoperative ultrasonography and MRI, mainly improving targeting lesions of eloquent cortex (e.g., motor cortex, or speech area). Moreover, procedures such as minimally invasive surgery (to selected locations such as base of skull) or awake craniotomy (with cortical mapping to assist in identifying vital area) are done to reduce the risk of complications. With advances in stereotactic and minimally invasive image guidance, other techniques have been developed such as laser interstitial thermal therapy for treatment of resistant brain metastases or radiation necrosis. Increasing use of SRS for larger lesions has led to fewer resections of brain metastases that were once considered too large for single-fraction SRS or that involve eloquent cortex. However, currently, there are no prospective data to evaluate the efficacy of these treatments compared with surgery.

Radiation Therapy

Whole-Brain Radiotherapy

Radiation therapy for the treatment of brain metastases has changed dramatically over the past several decades. In the past, the standard treatment for brain metastases was WBRT covering the entire intracranial contents ( Fig. 55.2A ). The benefits of WBRT were first described in the 1950s and 1960s. In these early studies, significant symptomatic improvement was noted in about 60% of patients, and the median survival time ranged from 3 to 6 months compared with an expected median survival time of 1 to 2 months without treatment. Moreover, WBRT was widely available, easily started without complicated planning, treats both gross and subclinical disease, and provides palliation for symptoms. All these made WBRT the standard of care for brain metastases for many years.

Figure 55.2, (A) A double-exposed portal image of a typical whole-brain radiotherapy field showing radiation covering the entire brain. The patient's eyes and other extracranial structures are blocked (right) . (B) Postcontrast T1-weighted magnetic resonance imaging of a brain metastasis shown on the day of radiosurgery with superimposed 50% and 25% isodose contours. A dose of 17.5 Gy was prescribed at the 50% isodose contour (left) .

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here