Open access

Medical Management of Brain Metastases from Lung Cancer

Written By

Ryuya Yamanaka

Submitted: 11 November 2010 Published: 23 August 2011

DOI: 10.5772/20980

From the Edited Volume

Brain Tumors - Current and Emerging Therapeutic Strategies

Edited by Ana L. Abujamra

Chapter metrics overview

3,353 Chapter Downloads

View Full Metrics

1. Introduction

Brain metastases are a frequent complication in patients with lung cancer and a significant cause of morbidity and mortality. Brain metastases are found in about 10-25% of patients at the time of diagnosis, and approximately 40-50% of all patients with lung cancer develop brain metastases during the course of their disease, with greater frequency at autopsy (approximately 50%) than predicted from the presence of symptoms (1). The incidence of brain metastasis is increasing mainly due to longer patient survivals resulting from newer treatment modalities. Most patients with lung cancer metastatic to the brain have multiple lesions (2). Brain metastases are usually associated with poor outcomes and shortened survival of 3 to 6 months. Standard treatment options include symptomatic therapy with corticosteroids and whole-brain radiotherapy (WBRT) (3), and more aggressive approaches such as surgery or radiosurgery are indicated in a subset of patients (4,5). Surgical resection of accessible brain metastases combined with postoperative WBRT is the management of choice for a single metastasis (6). However, radiosurgery for brain metastases produces high rates of tumor control similar to the rates obtained by excisional surgery (7). Patients with multiple brain metastases are commonly treated with WBRT for the palliation of symptoms (8). The role of radiosurgery for multiple brain metastases is less clear, but it can be effective (9). The poor outcomes and relapses following WBRT alone indicate a need for new therapeutic options. Generally, poor prognosis occurs not from cerebral problems, but from extracranial metastases, and death is caused by systemic disease combined with the neurological condition (10). However, treatment with systemic chemotherapy is controversial because chemotherapeutic agents may not cross the blood-brain-barrier (BBB) and therefore are less effective against central nervous system (CNS) disease than against extracranial, systemic disease. However, the BBB is partially disrupted in brain metastases (11) and similar concentrations of chemotherapeutic agents are found in intracerebral and extracerebral tumors (5). Brain metastases resulting from both non-small-cell (NSCLC) and small-cell lung cancer (SCLC) are susceptible to systemic chemotherapy, and cerebral response rates up to 50% were observed even in second-line treatment of NSCLC and SCLC (1,10,11). Still, medical therapies for brain metastases are neither well-studied nor established. Here, I analyze the impact of medical treatment on survival by reviewing recent articles and provide recommendations for the management of patients with brain metastases from lung cancer.

Advertisement

2. Non-small cell lung cancer (NSCLC)

NSCLC accounts for approximately 75% of lung cancer cases, with the majority of patients having inoperable, locally advanced or metastatic disease at the time of diagnosis, reflected in the low 5-year survival rate for all stages (currently 13%) (12). Despite two decades of cisplatin-based chemotherapy of advanced NSCLC, the survival benefit remains modest (13). New chemotherapy combinations have minimal impact on survival compared with older regimens, with overall response rates of approximately 30%, median survival benefits of 8–9 months, and 1-year survival rates of approximately 30% (14). New therapies are required that are effective against locally advanced or metastatic NSCLC.

2.1. Front-line chemotherapy (Table 1)

Many chemotherapeutic regimens have been tested in phase II or III trials for the treatment of brain metastases arising from NSCLC. There are 8 larger reports (15-22) with more than 10 patients, published from 1994 to 2008 in English, on front-line chemotherapy of brain metastases from NSCLC. In patients with NSCLC, 17-50% of patients with previously untreated brain metastases responded to a combination of cisplatin plus fotemustine; carboplatin plus etoposide; cisplatin plus teniposide; cisplatin plus etoposide; cisplatin plus ifosfamide, CPT-11; cisplatin plus vinorelbine; carboplatin plus vinorelbine, gemcitabine; cisplatin plus paclitaxel. Systemic disease activity correlates well with activity against brain metastases, but overall survival (OS) is still 4-12 months.

Author Regimen No. of patients Study design Response rate (%) Disease stabilization (%) mPFS (Month) OS (Month)
Cotto C CDDP,Fotemustine 31 Phase II 23 51.6 5 4
Malacarne P CBDCA,VP16 18 Phase II 17 39 n.d. 7.5
Minotti V CDDP,Teniposide 23 Phase II 35 65 7 5
Franciosi V CDDP,VP-16 43 Phase II 30 65 4 8
Fujita A CDDP,IFOS,CPT-11 28 Phase I/II 50 96 4.6 12
Robinet G CDDP,VNR 76 Phase III 27 n.d. 3.2 6
Bernardo G CBDCA,VNR,GEM 22 Phase II 45 85 6.2 8.2
Cortes J CDDP,Paclitaxel 26 Phase II 38 69 3.2 5.3

Table 1.

CBDCA,carboplatin; CDDP, cisplatin; CPT-11, irinotecan; GEM, gemcitabine; IFOS, ifosfamide; mOS, median overall survival; mPFS, median progression free survival; n.d., not determined; VNR, vinorelbine; VP16, etoposide. Modified from: Yamanaka R. Oncol Rep 2009;22:1269-1276 with permission from Spandidos Publications

2.2. Second-line chemotherapy

2.2.1. Epidermal growth factor receptor tyrosine kinase Inhibitor

The epidermal growth factor receptor (EGFR) is expressed in a variety of tumors, including NSCLC (23), and elevated EGFR expression is associated with a poor prognosis in lung cancer patients (24). Several EGFR-targeted agents have been developed, including gefitinib (ZD1839; Iressa) and erlotinib (OSI-774; Tarceva), an orally active anilinoquinazoline compound that inhibits EGFR tyrosine kinase activity (25). In two large, well-designed phase II clinical trials, refractory patients with NSCLC experienced overall response rates of 11.8–18.4%, median survival benefits of 6.5–7.6 months, and 1-year survival rates of 29–35% (26,27), with encouraging response rates in select patients (women, non-smokers, patients with adenocarcinoma, and specific EGFR mutations in the kinase domain) (28-33). Although targeting EGFR-associated tyrosine kinase with gefitinib and erlotinib results in durable responses in some patients, the activity of these drugs against brain metastases has been poorly documented so far.

Gefitinib (Table 2)

Phase II studies of gefitinib on brain metastases from NSCLC indicated objective responses occur in 33% of patients (Asia) (34,35) or 9.7% of patients (Europe) (36). In comparison, WBRT with 30 to 40 Gy for brain metastases from NSCLC results in objective responses in 38% to 45% of patients (37,38). Gefitinib is well tolerated, mostly with grade 1/2 skin rashes. The severity of skin toxicity was tightly associated with tumor response and patient survival (34). Gefitinib was most effective at treating brain metastases in patients with EGFR mutations in the tyrosine kinase domain (deletion mutation in two patients and a point mutation in one patient) in one study (39). However, this analysis was performed using tissue samples from primary lung cancer and not from metastatic brain lesions. Yokouchi et al (40). reported that some patients who experienced disease progression after responding to gefitinib were sensitive to gefitinib re-administration after temporary cessation of gefitinib and other treatments. Patients may still be expected to have prolonged survival if they once responded to gefitinib and then underwent various subsequent treatments followed by re-administration of gefitinib. These findings might provide valuable information for the management of gefitinib-responders. Although the survival benefit is controversial, gefitinib may also be useful for the treatment of carcinomatous meningitis from NSCLC to improve neurological dysfunction (41,42,43). Thus, gefitinib has therapeutic potential for palliative therapy in patients with brain metastases.

Author No. of patients Study design Previous WBRT/Chemo (%) Response rate (%) Disease stabilization (%) mPFS (Month) OS (Month)
Ceresoli GL 41 phase II 43.9/90.2 9.7 26.8 3 5
Chiu CH 76 phase II n.d./84.2 33.3 63.2 5 9.9
Wu C 40 phase II 65/100 32 77 9 15

Table 2.

Chemo, chemotherapy; mOS, median overall survival; mPFS, median progression free survival; n.d., not determined; WBRT, whole brain radiotherapy. Modified from: Yamanaka R. Oncol Rep 2009;22:1269-1276 with permission from Spandidos Publications.

Erlotinib

Erlotinib treatment of brain metastases from NSCLC has been reported in 20 cases (44-55). These were histologically confirmed as adenocarcinoma in the primary site. The main adverse events were Grade 1 skin rashes. Some patients had responses longer than 6 months. Erlotinib responses are higher in patients with a somatic mutation in EGFR or a point mutation in the activation loop of the kinase domain (56,57). Thirteen patients showed a response to erlotinib after gefitinib failure. Although gefitinib failure may result from cross-resistance to other EGFR tyrosine kinase inhibitors (EGFR-TKI), these cases suggest that re-challenging patients with EGFR-TKI may be beneficial. At the recommended dosage, erlotinib showed higher blood concentrations than gefitinib, and may also have higher in cerebral spinal fluid. In addition, two patients with intracranial lesions responded to erlotinib treatment although extra-cranial lesion progressed. In the Rupport case (51), a secondary T790M mutation associated with resistance to EGFR-TKI was found in the liver biopsy. Erlotinib was reintroduced and produced quick neurological improvement, even though the extra-cranial disease remained resistant to erlotinib. These cases also highlight the oligoclonal nature of NSCLC and its differential sensitivity to EGFR-TKIs, in that extra-cranial disease was resistant to erlotinib both initially and on re-challenge. Persistent cerebral TKI sensitivity should be considered in patients presenting with a CNS relapse after stopping EGFR-TKI, even with a T790M resistant mutation in non-cerebral metastases. In addition, erlotinib should be considered for treatment of intra-cranial disease.

2.2.2. Temozolomide (Table 3)

Temozolomide is an orally administered prodrug that is converted spontaneously to the active alkylating agent, monomethyl triazenoimidazole carboxamide, at physiologic pH, crosses the BBB, and has antitumor activity against malignant glioma, melanoma, NSCLC, and carcinoma of the ovary and colon (58). CNS concentrations reach approximately 30–40% of plasma levels, achieving therapeutic concentrations in the brain (59), and clearance of temozolomide is unaffected by co-administration with anticonvulsants, antiemetics, or dexamethasone (58,59). The dose-limiting toxicity is non-cumulative myelosuppression that rarely requires treatment delays or dose reductions. In patients with newly-diagnosed brain metastases or with progression after radiotherapy, temozolomide produces objective response rates between 5 and 10% (60-62) and is well tolerated.

Author Regimen No. of patients Study
design
Previous WBRT/Chemo (%) Response
rate (%)
Disease stabilization (%) mPFS (Month) OS (Month)
Giorgio TMZ 30 phase II 100/100 10 20 3.6 6
Kouroussis C TMZ 12 phase II n.d./100 8.3 25 n.d. n.d.
Abrey LE TMZ 22 phase II 100/n.d. 9 45 n.d. n.d.
Christodoulou
C
TMZ 12 phase II 100/n.d. 8 n.d. n.d. n.d.

Table 3.

TMZ,temozolomide; Chemo, chemotherapy; mOS, median overall survival; mPFS, median progression free survival; n.d., not determined; WBRT, whole brain radiotherapy. Modified from: Yamanaka R. Oncol Rep 2009;22:1269-1276 with permission from Spandidos Publications

2.2.3. Temozolomide plus other chemotherapeutic agents

Preclinical experiments and early clinical studies in other malignancies indicate that temozolomide may have additive or synergistic effects when used with other chemotherapeutic agents (64,65). In addition, its minimal toxicity allows for the combination of temozolomide with gemcitabine, gemcitabine/cisplatin, or gemcitabine/vinorelbine in NSCLC patients to produce dramatic cerebral responses (66,67). The combination of temozolomide with other chemotherapeutic agents represents a promising strategy for treating brain metastases.

2.3. Chemotherapy plus whole-brain irradiation (Table 4)

Combining chemotherapy with brain radiotherapy is attractive because chemotherapy is active against both primary tumors and brain metastases, and because chemotherapy may act as a radiosensitizer. Two studies have compared randomized chemotherapy alone with chemotherapy/WBRT. Quantin et al. (68) reported a phase II study of radiotherapy plus vinorelbine, ifosfamide, and cisplatin chemotherapy in patients with brain metastases of NSCLC. The response rate was 56% and median survival was 7.6 months. The same author also reported a phase II study with concomitant brain radiotherapy and high-dose ifosfamide in brain relapses (69). Median survival was 13 months. Myelosuppression was the main toxic effect, but remained manageable and no toxic deaths occurred. The high response rate for brain lesions and improvement in neurological symptoms deserves further exploration.

Author Chemotherapy Regimen No. of patients Study
design
Previous WBRT/Chemo (%) Response rate (%) Disease stabilization (%) mPFS (Month) OS (Month)
Quantin CDDP,VNR, IFOS 23 phase II 0/0 56 65 n.d. 7.6
Quantin high-dose
IFOS
16 phase II n.d./n.d. 25 n.d. n.d. 13
Ma S Gefitinib 25 phase II 0/0 81 95.2 10 13
Addeo R TMZ 15 phase II 44/74 6.5 60 6 8.8
Cortot CDDP,TMZ 50 phase II 6/n.d. 12 54 2.3 5
Robinet CDDP,VNR 85 phase III 0/0 28 n.d. 2.7 5.2

Table 4.

CDDP, cisplatin; Chemo, chemotherapy; IFOS, ifosfamide; mOS, median overall survival; mPFS, median progression free survival; n.d., not determined; TMZ,temozolomide; VNR,vinorelbine; WBRT, whole brain radiotherapy. Modified from: Yamanaka R. Oncol Rep 2009;22:1269-1276 with permission from Spandidos Publications

Ma et al. (70) found that treatment with concomitant gefitinib and WBRT was well tolerated, with significant improvement of neurological symptoms in a Chinese population with brain metastases from NSCLC. Addeo et al. (71) reported response rates of 6.5% using a combination of temozolomide and WBRT. Cortot et al. (72) reported response rates of 12% with temozolomide, cisplatin, and WBRT. A randomized phase II study evaluated the efficacy of concurrent temozolomide and radiotherapy versus radiotherapy alone in 58 patients with previously untreated brain metastases from different solid tumors (31 patients had NSCLC) (73). The temozolomide group showed significant improvements in cerebral response rate (96% versus 67%), and temozolomide was safe and well-tolerated. However, overall survival rates and changes in neurological function were similar in both groups. Robinet et al. (74) reported a phase III study comparing the timing of WBRT, either before or after chemotherapy, and found a 28% response rate in 85 patients treated with cisplatin and vinorelbine in the early WBRT arm. The median survival in this arm was 5.2 months and median time to progression (TTP) was 2.1 months. Radiotherapy timing did not change survival time. Thus, for NSCLC, WBRT should be added to initial chemotherapy if there is no treatment response.

Advertisement

3. Small Cell Lung Cancer

The brain is the most common metastatic site in SCLC, and is usually fatal. Approximately 18-25% of SCLC patients have brain metastases already at diagnosis, and an additional 50% will develop CNS involvement during their disease course (75-77). Although WBRT and corticosteroids are the treatment of choice, systemic chemotherapy may also have therapeutic value. Extracranial disease is almost always present in SCLC, and chemotherapy can treat both brain metastases and these other disease sites. Prophylactic cranial irradiation (PCI) for patients responsive to induction therapy markedly reduces the risk of CNS relapse and significantly improves survival (77,78). Surgical treatment for solitary lesions or systemic chemotherapy for multifocal brain metastases that are minimally symptomatic can be useful, particularly when these patients also have extracranial metastatic disease. Thus, systemic chemotherapy can complement WBRT for treatment of brain metastases in SCLC.

3.1. Front-line chemotherapy (Table 5)

There are 4 larger reports (79-82) with more than 10 patients, published from 1989 to 2008 in English, on front-line chemotherapy of brain metastases from SCLC. The chemotherapeutic regimens, including cyclophosphamide, vincristine, etoposide, doxorubicin and cisplatin, produced response rates of 27-82% Thus, chemotherapy followed by radiation therapy may be first-line treatment for patients with systemic disease and asymptomatic brain metastases.

Author Regimen No.
of patients
Study
design
Response
rate (%)
Disease
stabilization (%)
mPFS
(Month)
OS (Month)
Lee JS CTX,DX,VCR,VP-16 11 Phase II 82 91 6 8.5
Twelves CJ CTX,VP-16,VCR 25 Retrospective 53 n.d. 5.5 8.5
Kristjansen PE CDDP,VP-16,VCR 21 Phase II 52 57 4.5 3.7
Seute T CTX,DX,VP-16 22 Phase II 27 50 n.d. n.d.

Table 5.

CTX, cyclophosphamide; DX, doxorubicin; mOS, median overall survival; mPFS, median progression free survival; n.d., not determined; VCR, vincristine; VP16, etoposide; Modified from: Yamanaka R. Oncol Rep 2009;22:1269-1276 with permission from Spandidos Publications.

3.2. Second-line chemotherapy (Table 6)

SCLC relapse may also require systemic chemotherapy, which showed efficacy in 7 small phase II studies (83-89) with chemotherapy as salvage treatment after failing systemic chemotherapy and WBRT. Response rates are generally lower and survival is decreased in patients who receive chemotherapy for brain metastases after failure following radiotherapy. Postmus et al. (83) reported a response rate of 43% in the brain after high-dose etoposide. Groen et al. (84) reported a response rate of 40% with carboplatin and Postmus et al. (85) reported a response rate of 42% with a single agent, teniposide. The response rates of the primary tumor are not given in these reports. Chen et al. (89) reported a high response rate of 65% with a combination of carboplatin and irinotecan. In an analysis by Schuette et al. (86) and Korfel et al. (88), response rates for brain metastases of 50% and 33%, respectively, were achieved with topotecan. In both, the cerebral response rate was superior to the response rate of the primary tumor, probably because the intact BBB during the first treatment round protected tumor cells of the brain metastases. However, the severe adverse events associated with these regimens would be difficult to tolerate for pretreated patients who had already received radiation and multiple regimens of myelosuppressive chemotherapy. Treatment-related mortality was observed in 7 of 13 patients treated with high-dose etoposide (83) and in 8 of 80 patients treated with teniposide (85).

Author Regimen No. of patients Study design Response rate (%) Disease stabilization (%) mPFS (Month) OS (Month)
Postmus PE HD-VP16 23 Phase II 43 52 n.d. 8
Groen H CBDCA 20 Phase II 40 60 2 4
Postmus PE Teniposide 80 Phase II 33 47.5 4.8 2.9
Schuette W Topotecan 22 Phase II 50 82 n.d. 6
Postmus PE Teniposide 60 Phase III 22 43 4.5 3.2
Korfel A Topotecan 30 Phase II 33 60 3.1 3.6
Chen G CBDCA, CPT-11 15 Phase II 65 86 n.d. 6

Table 6.

CBDCA,carboplatin; CPT-11, irinotecan; mOS, median overall survival; mPFS, median progression free survival; n.d., not determined; HD-VP16, high dose etoposide Modified from: Yamanaka R. Oncol Rep 2009;22:1269-1276 with permission from Spandidos Publications

Temozolomide shows low response rates when used alone. Ebert et al. (90) reported a case of a patient with SCLC with recurrent brain metastases who was treated with temozolomide and oral etoposide. This regimen was well tolerated and resulted in dramatic, durable responses. Combining temozolomide with other chemotherapeutic agents represents a promising strategy for treating patients with brain metastases from SCLC.

3.3. Chemotherapy plus whole-brain irradiation

Postmus et al. (87) reported a phase III study where 120 SCLC patients with brain metastases were randomized to receive teniposide with or without WBRT. Combined treatment produced a 57% response rate, and teniposide alone produced a 22% response rate. Combined treatment produced a longer TTP, but both regimens produced similar clinical responses outside the brain, median survival times (median survival 3.5 and 3.2 months, respectively) and symptomatic improvement. Further studies are needed to compare combinations of WBRT with chemotherapy.

Advertisement

4. Discussion

The impairment of physical, cognitive, and affective function that accompanies most brain metastases is highly distressing and can be seen as a "loss" of the patient even before death. Improved treatment of overt brain metastases may have palliative value and eradication of microscopic brain disease may cure patients already cured in other sites. Assumptions about BBB penetration and chemotherapy resistance have limited the use of chemotherapy for treatment of brain metastases. Small, lipid-soluble molecules can penetrate the normal BBB barrier, but large, hydrophilic molecules cannot. Furthermore, high levels of the multidrug transporter, P-gp, are expressed in the endothelial cells of brain capillaries. P-gp actively prevents drugs from passing through the BBB (91). However, macroscopic metastases, relapsed disease, and radiation therapy can disrupt the BBB (92), as shown via magnetic resonance imaging (MRI) or computed tomography (CT) of intravenous contrast inside intracerebral lesions. In addition, the concentration of chemotherapy drugs, including platinum, is similar in intracerebral and extracerebral tumors (5). Cytostatics unable to penetrate the BBB produce comparable response rates for cerebral metastases and systemic disease, and adding BBB-penetrating drugs such as procarbazine, nitrosoureas, or methotrexate to a standard combination regimen did not improve the CNS relapse frequency (93,94). The chemosensitivity of the primary tumor is the major determinant of the response to systemic treatment for brain metastases (92,95), although asymptomatic brain metastases may have lower responses than systemic tumors to systemic chemotherapy (82).

Dexamethasone and enzyme-inducing anti-epileptic drugs (EIAEDs) can induce cytochrome p450 3A isoenzymes, including CYP3A4, which metabolizes chemotherapeutic agents (96,97) including paclitaxel, irinotecan, vinorelbine, cyclophosphamide, doxorubicin, etoposide, ifosfamide, teniposide, erlotinib, and gefitinib. Therefore, co-administration of EIAEDs or dexamethasone may increase the metabolism of chemotherapeutic agents, lower plasma concentrations, and reduce efficacy.

Response and survival rates are generally lower after chemotherapy for brain metastases following radiotherapy failure (84). Combination regimens also produce side effects that would be difficult to tolerate after radiation or multiple regimens of myelosuppressive chemotherapy. Oral agents such as gefitinib, erlotinib, and temozolomide were well tolerated even in pretreated patients, confirming their favorable adverse event profile. In a molecularly selected population with brain metastases, these agents can produce high response rates.

Brain metastases resulting from both NSCLC and SCLC are susceptible to systemic chemotherapy, with cerebral response rates similar to primary tumor responses, even in second-line treatment. Clinical conditions such as a chemotherapy-sensitive primary tumor, no prior chemotherapy, or the presence of systemic metastases should indicate the use of chemotherapy. The brain is rarely the sole site of metastases in lung cancer, and patients receiving cranial irradiation alone often die of extra-cranial tumors rather than cerebral metastases. Chemotherapy can control other disease sites and is generally better tolerated than WBRT. Chemotherapy should be initiated before WBRT because chemotherapy cannot be given for 1 month after WBRT and concomitant WBRT/chemotherapy is more toxic. Combinations of these therapeutic modalities for the management of brain metastases require further testing in randomized phase III studies. Because of the short survival times, the late effects of cranial irradiation such as dementia may be underestimated because they do not usually present until months or years after treatment. Kristensen et al. (98) showed response rates of 76% in primary brain metastases from SCLC but only 43% in relapsed metastases, similar to other SCLC metastatic sites. Chemotherapy has a clearer therapeutic impact in SCLC than NSCLC. Thus, chemotherapy should be incorporated into the management of brain metastases as part of a multimodal treatment concept.

First-line chemotherapy can be performed in patients with asymptomatic or minor neurological symptoms or other metastatic sites, as well as for relapses after radiotherapy or systemic chemotherapy. The main goal of cytostatic therapy is palliation, with clinical improvement and brief, limited duration of high-dose steroid treatments critical to this palliation. The inclusion of patients with brain metastases from lung cancer in prospective trials of new therapeutic agents or combinations should be pursued.

References

  1. 1. CA Kristensen Kristjansen. P. E. Hansen H. Systemic chemotherapy. of brain. metastases from. small-cell lung. cancer a. review J Clin Oncol 10 1992 9 1498 1502
  2. 2. Wen P. Y. Mc Laren Black. P. Loeffler J. S. Metastatic brain. cancer In. Cancer Principles. Practice of. Oncology 6th. edition De Vita VT Jr, Hellman S, Rosenberg SA (eds.) Lippincott Williams & Wilkins, Philadelphia, PA, 2655 2670 1999
  3. 3. Lagerwaard FJ, Levendag PC, Nowak PJCM, et al: Identification of prognostic factors in patients with brain metastases: a review of 1292 patients.Int J Radiat Oncol Biol Phys 43 1999 795 803
  4. 4. Sheenan J. P. Sun M. H. Kondziolka D. et al. Radiosurgery for. non-small cell. lung carcinoma. metastatic to. the brain. long-term outcomes. prognostic factors. influencing patient. survival time. local tumor. control JP, Sun MH, Kondziolka D, et al: Radiosurgery for non-small cell lung carcinoma metastatic to the brain: long-term outcomes and prognostic factors influencing patient survival time and local tumor control. J Neurosurg 97 2002 1276 1281
  5. 5. Stewart D. J. Leavens M. Maor M. et al. Human central. nervous system. distribution of. cis-diamminedichloroplatinum as use a. radiosensitizer in. malignant brain. tumors Cancer Res 42 1982 6 2474 2479
  6. 6. Patchell R. A. Tibbs P. A. Walsh J. W. et al. A. randomized trial. of surgery. in the. treatment of. single metastases. to the. brain N Engl J Med 322 1990 8 494 500
  7. 7. Kihlström L. Karlsson B. Lindquist C. Gamma Knife. surgery for. cerebral metastases. Implications for survival based on 16 years experience. Stereotact Funct Neurosurg 61 Suppl 1 1993 45 50
  8. 8. Horton J. Baxter D. H. Olson K. B. The management. of metastases. to the. brain by. irradiation corticosteroids Am J Roentgenol Radium Ther Nucl Med 111 1971 2 334 336
  9. 9. Fukuoka S. Seo Y. Takanashi M. et al. Radiosurgery of. brain metastases. with the. Gamma Knife. Stereotact Funct Neurosurg 66 Suppl 1 1996 193 200
  10. 10. Siegers HP: Chemotherapy for brain metastases: recent developments and clinical considerations.Cancer Treat Rev 17 1990 1 63 76
  11. 11. Lesser GJ: Chemotherapy of cerebral metastases from solid tumors.Neurosurg Clin N Am 7 1996 3 527 536
  12. 12. Stanley K. Stjernsward J. Lung cancer-a. worldwide health. problem Chest 96 Suppl 1:1S-5S, 1989
  13. 13. Non-small Cell Lung Cancer Collaborative Group: Chemotherapy in non-small cell lung cancer: a meta-analysis using updated data on individual patients from 52 randomised clinical trials.BMJ 311 1995 899 909
  14. 14. Schiller J. H. Harrington D. CP Belani et. al Eastern. Cooperative Oncology. Group Comparison of four chemotherapy regimens for advanced non-small-cell lung cancer. New Engl J Med 346 2002 92 98
  15. 15. Cotto C. Berille J. Souquet P. J. et al. A. phase I. I. trial of. fotemustine cisplatin in. central nervous. system metastases. from non-small. cell lung. cancer Eur J Cancer 32A(1):69 EOF 71 EOF 1996
  16. 16. Malacarne P. Santini A. Maestri A. Response of. brain metastases. from lung. cancer to. systemic chemotherapy. with carboplatin. etoposide Oncology 53 1996 3 210 213
  17. 17. Minotti V. Crinò L. Meacci M. L. et al. Chemotherapy with. cisplatin teniposide for. cerebral metastases. in non-small. cell lung. cancer Lung Cancer 20 1998 2 93 98
  18. 18. Franciosi V. Cocconi G. Michiara M. et al. Front-line chemotherapy. with cisplatin. etoposide for. patients with. brain metastases. from breast. carcinoma nonsmall. cell lung. carcinoma or. malignant melanoma. a. prospective study. Cancer 85 1999 7 1599 1605
  19. 19. Fujita A. Ohkubo T. Hoshino H. et al. Phase I. I. study of. cisplatin ifosfamide. irinotecan with. rh-C G. support S. F. in patients. with stage. I. I. Ib non-small-cell I. V. lung cancer. Br J Cancer 89 2003 6 1008 1012
  20. 20. Robinet G. Thomas P. Breton J. L. et al. Results of. a. phase I. I. I. study of. early versus. delayed whole. brain radiotherapy. with concurrent. cisplatin vinorelbine combination. in inoperable. brain metastasis. of non-small-cell. lung cancer. Groupe Français. de Pneumo-Cancérologie . G. F. P. C. Protocol 95-. Ann Oncol 12 2001 1 59 67
  21. 21. Bernardo G. Cuzzoni Q. Strada M. R. et al. First-line chemotherapy. with vinorelbine. gemcitabine carboplatin in. the treatment. of brain. metastases from. non-small-cell lung. cancer a. phase I. I. study Cancer Invest 20 2002 3 293 302
  22. 22. Cortes J. Rodriguez J. Aramendia J. M. et al. Front-line paclitaxel/cisplatin-based. chemotherapy in. brain metastases. from non-small-cell. lung cancer. Oncology 64 2003 1 28 35
  23. 23. Rusch V. Baselga J. Cordon-Cardo C. et al. Differential expression. of the. epidermal growth. factor receptor. its ligands. in primary. non-small cell. lung cancers. adjacent benign. lung Cancer Res 53 1993 2379 2385
  24. 24. Volm M. Rittgen W. Drings P. Prognostic value. of E. R. B. B- cyclin V. E. G. F. A. F. O. S. J. U. N. in M. Y. C. patients with. squamous cell. lung carcinomas. Br J Cancer 77 1998 663 669
  25. 25. DS Lawrence Niu J. Protein kinase. inhibitors the. tyrosine-specific protein. kinases Pharmacol Ther 77 1998 81 114
  26. 26. Fukuoka M. Yano S. Giaccone G. et al. Multi-institutional randomized. phase I. I. trial of. gefitinib for. previously treated. patients with. advanced non-small-cell. lung cancer. J Clin Oncol 21 2003 2237 2246
  27. 27. Kris MG, Natale RB, Herbst RS, et al: Efficacy of gefitinib, an inhibitor of the epidermal growth factor receptor tyrosine kinase, in symptomatic patients with non-small cell lung cancer.A randomized trial. JAMA 290 2003 2149 2158
  28. 28. Fukuoka M. Yano S. Giaccone G. et al. Multi-institutional randomized. phase I. I. trial of. gefitinib for. previously treated. patients with. advanced non-small-cell. lung cancer. . The I. D. E. A. L. . Trial J Clin Oncol 21 2003 12 2237 2246
  29. 29. Kris MG, Natale RB, Herbst RS, et al: Efficacy of gefitinib, an inhibitor of the epidermal growth factor receptor tyrosine kinase, in symptomatic patients with non-small cell lung cancer: a randomized trial.JAMA 290 2003 16 2149 2158
  30. 30. MS Tsao Sakurada. A. Cutz J. C. et al. Erlotinib in. lung-molecular cancer. clinical predictors. of outcome. N Engl J Med 353 2005 2 133 144
  31. 31. Cappuzzo F. Hirsch F. R. Ross E. et al. Epidermal growth. factor receptor. gene protein gefitinib sensitivity. in non-small-cell. lung cancer. J Natl Cancer Inst 97 2005 9 643 655
  32. 32. Paez JG, Janne PA, Lee JC, et al: EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy.Science 304 2004 5676 1497 1500
  33. 33. Eberhard DA,Johnson BE, Amler LC, et al: Mutations in the epidermal growth factor receptor and in KRAS are predictive and prognostic indicators in patients with non-small-cell lung cancer treated with chemotherapy alone and in combination with erlotinib. J Clin Oncol 23 2005 25 5900 5909
  34. 34. Chiu CH, Tsai CM, Chen YM, et al: Gefitinib is active in patients with brain metastases from non-small cell lung cancer and response is related to skin toxicity.Lung Cancer 47 2005 1 129 138
  35. 35. Wu C. Li Y. L. Wang Z. M. et al. as Gefitinib palliative. therapy for. lung adenocarcinoma. metastatic to. the brain. Lung Cancer 57 2007 3 359 364
  36. 36. Ceresoli G. L. Cappuzzo F. Gregorc V. et al. Gefitinib in. patients with. brain metastases. from non-small-cell. lung cancer. A. prospective trial. Ann Oncol 15 2004 1042 1047
  37. 37. Antoniou D. Kyprianou K. Stathopoulos G. P. et al. Response to. radiotherapy in. brain metastases. survival of. patients with. non-small cell. lung cancer. Oncol Rep 14 2005 733 736
  38. 38. Nieder C. Niewald M. Hagen T. Brain metastases. in bronchial. breast carcinoma. Differences in. metastatic behavior. prognosis . German Radiologe 35 1995 816 821
  39. 39. Shimato S. Mitsudomi T. Kosaka T. et al. E. G. F. R. mutations in. patients with. brain metastases. from lung. cancer association. with the. efficacy of. gefitinib Neuro Oncol 8 2006 2 137 144
  40. 40. Yokouchi H. Yamazaki K. Kinoshita I. et al. Clinical benefit. of readministration. of gefitinib. for initial. gefitinib-responders with. non-small cell. lung cancer. B. M. BMC Cancer 7:51 EOF 2007
  41. 41. Sakai M, Ishikawa S, Ito H, et al: Carcinomatous meningitis from non-small-cell lung cancer responding to gefitinib.Int J Clin Oncol 11(3):243-245, 2006.
  42. 42. Kim MK, Lee KH, Lee JK, et al: Gefitinib is also active for carcinomatous meningitis in NSCLC.Lung Cancer 50 2005 2 265 269
  43. 43. So T. Inoue M. Chikaishi Y. et al. Gefitinib ventriculo-peritoneal a. shunt to. manage carcinomatous. meningitis from. non-small-cell lung. cancer report. of two. cases Surg Today 39 2009 7 598 602
  44. 44. Lai C. S. Boshoff C. Falzon M. Lee S. M. Complete response. to erlotinib. treatment in. brain metastases. from recurrent. N. S. C. L. C. Thorax 61(1):91 EOF 91 EOF 2006
  45. 45. Popat S, Hughes S, Papadopoulos P, et al: Recurrent responses to non-small cell lung cancer brain metastases with erlotinib.Lung Cancer 56(1):135-137,2007.
  46. 46. Chang JW, Chou CL, Huang SF, et al: Erlotinib response of EGFR-mutant gefitinib-resistant non-small-cell lung cancer.Lung Cancer 58 2007 3 414 417
  47. 47. Gounant V. Wislez M. Poulot V. et al. Subsequent brain. metastasis responses. to epidermal. growth factor. receptor tyrosine. kinase inhibitors. in a. patient with. non-small-cell lung. cancer Lung Cancer 58 2007 3 425 428
  48. 48. Fekrazad M. H. Ravindranathan M. Jones D. V. Jr:  Response. of intracranial. metastases to. erlotinib therapy. Jones DV Jr:  Response of intracranial metastases to erlotinib therapy. J Clin Oncol 25 2007 31 5024 5026
  49. 49. von Pawel. J. Wagner H. Duell T. Poellinger B. Erlotinib in. patients with. previously irradiated. recurrent brain. metastases from. non-small cell. lung cancer. two case. reports Onkologie31 2008 3 123 126
  50. 50. Altavilla G. Arrigo C. Santarpia M. C. et al. Erlotinib therapy. in a. patient with. non-small-cell lung. cancer brain metastases. J Neurooncol 90 2008 1 31 33
  51. 51. Ruppert A. M. Beau-Faller M. Neuville A. et-T al. E. G. F. R. K. I. lung adenocarcinoma. with C. N. S. relapse interest. of molecular. follow-up Eur Respir J 33 2009 2 436 440
  52. 52. Katayama T. Shimizu J. Suda K. et al. Efficacy of. erlotinib for. brain leptomeningeal metastases. in patients. with lung. adenocarcinoma who. showed initial. good response. to gefitinib. J, Suda K, et al: Efficacy of erlotinib for brain and leptomeningeal metastases in patients with lung adenocarcinoma who showed initial good response to gefitinib. J Thorac Oncol 4 2009 11 1415 1419
  53. 53. Kunimasa K. Yoshioka H. Iwasaku M. et al. Two cases. of non-small. cell lung. cancer which. developed central. nervous system. metastases during. gefitinib treatment. improving after. changing to. erlotinib treatment. Nihon Kokyuki Gakkai Zasshi 48 2010 2 166 171
  54. 54. Fujikura Y. Morishima Y. Ota K. et al. Satisfactory outcome. with erlotinib. after failure. with gefitinib. in a. patient with. meningeal carcinomatosis. secondary to. non-small cell. lung cancer. Nihon Kokyuki Gakkai Zasshi 48 2010 5 391 396
  55. 55. Clarke J. L. Pao W. Wu N. et al. High dose. weekly erlotinib. achieves therapeutic. concentrations in. C. S. F. is effective. in leptomeningeal. metastases from. epidermal growth. factor receptor. mutant lung. cancer JL, Pao W, Wu N, et al: High dose weekly erlotinib achieves therapeutic concentrations in CSF and is effective in leptomeningeal metastases from epidermal growth factor receptor mutant lung cancer. J Neurooncol 99 2010 2 283 286
  56. 56. Pao W. Miller V. A. Epidermal growth. factor receptor. mutations small-molecule. kinase inhibitors. non-small-cell lung. cancer current. knowledge future directions. J Clin Oncol 23 2005 2556 2568
  57. 57. MS Tsao Sakurada. A. Cutz J. C. et al. Erlotinib in. lung cancer. molecular clinical predictors. of outcome. N Engl J Med 353 2005 133 144
  58. 58. Baker S. D. Wirth M. Statkevich P. et al. Absorption metabolism. excretion of. . C-temozolomide following. oral administration. to patients. with advanced. cancer Clin Cancer Res 5 1999 309 317
  59. 59. Ostermann S. Csajka C. Buclin T. et al. Plasma cerebrospinal fluid. population pharmacokinetics. of temozolomide. in malignant. glioma patients. Clin Cancer Res 10 2004 3728 3736
  60. 60. Giorgio C. G. Giuffrida D. Pappalardo A. et al. Oral temozolomide. in heavily. pre-treated brain. metastases from. non-small cell. lung cancer. phase I. I. study Lung Cancer 50 2005 2 247 254
  61. 61. Kouroussis C. Vamvakas L. Vardakis N. et al. Continuous administration. of daily. low-dose temozolomide. in pretreated. patients with. advanced non-small. cell lung. cancer a. phase I. I. study Oncology 76 2009 2 112 117
  62. 62. Abrey L. E. JD Olson Raizer. J. J. et al. A. phase I. I. trial of. T. M. Z. for patients. with recurrent. or progressive. brain metastases. JD, Raizer JJ, et al: A phase II trial of TMZ for patients with recurrent or progressive brain metastases. J Neurooncol 53 2001 259 265
  63. 63. Christodoulou C. Bafaloukos D. Kosmidis P. et al. Phase I. I. study of. temozolomide in. heavily pretreated. cancer patients. with brain. metastases Ann Oncol 12 2001 249 254
  64. 64. D’Atri S. Graziani G. Lacal P. M. et al. Attenuation of. O6-methylguanine-D N. A. methyltransferase activity. levels m. R. N. A. by cisplatin. temozolomide in. jurkat cells. J Pharmacol Exp Ther 294 2000 664 671
  65. 65. Piccioni D. D’Atri S. Papa G. et al. Cisplatin increases. sensitivity of. human leukemic. blasts to. triazene compounds. J Chemother 7 1995 224 229
  66. 66. Ebert B. L. Niemerko E. Shaffer K. et al. Use of. temozolomide with. other cytotoxic. chemotherapy in. the treatment. of patients. with recurrent. brain metastases. from lung. cancer Oncologist 8 2003 69 75
  67. 67. Mangiameli A. Mineo G. Trovato G. Temozolomide . T. M. Z. in patients. with brain. metastases from. N. S. C. L. C. in combination. with brain. metastases from. N. S. C. L. C. in combination. with gemcitabine-cisplatin. . G. E. M. C. D. D. P. or gemcitabine-vinorelbine. . G. E. M. V. N. B. Proc Am Soc Clin Oncol 20:A2797,2001
  68. 68. Quantin X. Khial F. Reme-Saumon M. et al. Concomitant brain. radiotherapy vinorelbine-ifosfamide-cisplatin chemotherapy. in brain. metastases of. non-small cell. lung cancer. Lung Cancer 26 1999 25 39
  69. 69. Quantin X. Pujol J. L. Paris A. et al. Concomitant brain. radiotherapy high-dose ifosfamide. in brain. relapses of. lung cancer. Ann Oncol 8 1997 9 911 913
  70. 70. Ma Xu S. Deng Y. Yu Q. Treatment X. of brain. metastasis from. non-small cell. lung cancer. with whole. brain radiotherapy. Gefitinib in. a. Chinese population. Lung Cancer 65 2009 2 198 203
  71. 71. Addeo R. De Rosa C. Faiola V. et al. Phase . trial of. temozolomide using. protracted low-dose. whole-brain radiotherapy. for nonsmall. cell lung. cancer breast cancer. patients with. brain metastases. Cancer 113 2008 9 2524 2531
  72. 72. Cortot A. B. Gerinière L. Robinet G. et al. Phase I. I. trial of. temozolomide cisplatin followed. by whole. brain radiotherapy. in non-small-cell. lung cancer. patients with. brain-G metastases. a. G. L. O. T. study F. P. C. Ann Oncol 17 2006 9 1412 1417
  73. 73. Antonodau D. Paraskevaidis M. Sarris G. et al. Phase I. I. randomized trial. of temozolomide. concurrent radiotherapy. in patients. with brain. metastases J Clin Oncol 20 2002 3644 3650
  74. 74. Robinet G. Thomas P. Breton J. L. et al. Results of. a. phase I. I. I. study of. early versus. delayed whole. brain radiotherapy. with concurrent. cisplatin vinorelbine combination. in inoperable. brain metastases. of non-small. cell lung. cancer groupe. francais de pneumocancerologie. . G. F. P. C. protocol 95-. Ann Oncol 12 2001 59 67
  75. 75. van de Pol M. van Oosterhout A. G. Wilmink J. T. et al. M. R. I. in detection. of brain. metastases at. initial staging. of small-cell. lung cancer. Neuroradiology 38 1996 3 207 210
  76. 76. Seute T. Leffers P. ten Velde. G. P. Twijnstra A. Neurologic disorders. in 4. consecutive patients. with small. cell lung. carcinoma Cancer 100 2004 4 801 806
  77. 77. Komaki R. JD Cox Whitson. W. Risk of. brain metastasis. from small. cell carcinoma. of the. lung related. to length. of survival. prophylactic irradiation. Cancer Treat Rep 65(9-10):811 EOF 4 EOF 1981
  78. 78. Arriagada R. Le Chevalier T. Borie F. et al. Prophylactic cranial. irradiation for. patients with. small-cell lung. cancer in. complete remission. J Natl Cancer Inst 87 1995 3 183 190
  79. 79. LeeJS, Murphy WK, Glisson BS, et al: Primary chemotherapy of brain metastasis in small-cell lung cancer. J Clin Oncol 7 1989 7 916 922
  80. 80. Twelves CJ, Souhami RL, Harper PG, et al: The response of cerebral metastases in small cell lung cancer to systemic chemotherapy.Br J Cancer 61 1990 1 147 150
  81. 81. Kristjansen P. E. Soelberg Sørensen. P. Skov Hansen. M. Hansen H. H. Prospective evaluation. of the. effect on. initial brain. metastases from. small cell. lung cancer. of platinum-etoposide. based induction. chemotherapy followed. by an. alternating multidrug. regimen Ann Oncol 4 1993 7 579 583
  82. 82. Seute T. Leffers P. Wilmink J. T. et al. Response of. asymptomatic brain. metastases from. small-cell lung. cancer to. systemic first-line. chemotherapy JT, et al: Response of asymptomatic brain metastases from small-cell lung cancer to systemic first-line chemotherapy. J Clin Oncol 24 2006 13 2079 2083
  83. 83. Postmus P. E. Haaxma-Reiche H. Sleijfer D. T. et al. High-dose etoposide. for central. nervous system. metastases of. small cell. lung cancer. Preliminary results. Eur J Respir Dis Suppl 149 1987 65 71
  84. 84. Groen H. J. Smit E. F. Haaxma-Reiche H. Postmus P. E. as Carboplatin second. line treatment. for recurrent. or progressive. brain metastases. from small. cell lung. cancer Eur J Cancer 29A(12):1696 EOF 9 EOF 1993
  85. 85. Postmus P. E. Smit E. F. Haaxma-Reiche H. et al. Teniposide for. brain metastases. of small-cell. lung cancer. a. phase I. I. study European Organization for Research and Treatment of Cancer Lung Cancer Cooperative Group. J Clin Oncol 13 1995 3 660 665
  86. 86. Schütte W. Manegold C. von Pawel. J. V. et al. Topotecan--a new. treatment option. in the. therapy of. brain metastases. of lung. cancer Front Radiat Ther Oncol 33 1999 354 363
  87. 87. Postmus P. E. Haaxma-Reiche H. Smit E. F. et al. Treatment of. brain metastases. of small-cell. lung cancer. comparing teniposide. teniposide with. whole-brain radiotherapy--a. phase I. I. I. study of. the European. Organization for. the Research. Treatment of. Cancer Lung. Cancer Cooperative. Group J Clin Onol 18 2000 19 3400 3408
  88. 88. Korfel A. Oehm C. von Pawel. J. et al. Response to. topotecan of. symptomatic brain. metastases of. small-cell lung. cancer also. after whole-brain. irradiation awel J, et al: Response to topotecan of symptomatic brain metastases of small-cell lung cancer also after whole-brain irradiation. a multicentre phase II study. Eur J Cancer 38 2002 13 1724 1729
  89. 89. Chen G. Huynh M. Chen A. et al. Chemotherapy for. metastases in. small-cell lung. cancer Clin Lung Cancer 9 2008 1 35 38
  90. 90. Ebert B. L. Niemierko E. Shaffer K. Salgia R. Use of. temozolomide with. other cytotoxic. chemotherapy in. the treatment. of patients. with recurrent. brain metastases. from lung. cancer Oncologist 8 2003 1 69 75
  91. 91. Régina A. Demeule M. Laplante A. et al. Multidrug resistance. in brain. tumors roles. of the. blood brain. barrier Cancer Metastasis Rev 20 2001 13 25
  92. 92. Postmus P. E. Smit E. F. Chemotherapy for. brain metastases. of lung. cancer A. review Ann Oncol 10 1999 753 759
  93. 93. Bunn PA Jr, Nugent JL, Matthews MJ: Central nervous system metastases in small cell bronchogenic carcinoma.Semin Oncol 5 1978 3 314 322
  94. 94. Neijstrom ES, Capizzi RL, Rudnick SA, et al: High-dose methotrexate in small cell lung cancer.Lack of efficacy in preventing CNS relapse. Cancer 51 1983 6 1056 1061
  95. 95. van den Bent MJ: The role of chemotherapy in brain metastases.Eur J Cancer 39 2003 15 2114 2120
  96. 96. McCune JS, Hawke RL, LeCluyse EL, et al: In vivo and in vitro induction of human cytochrome P4503A4 by dexamethasone.Clin Pharmacol Ther 68 2000 4 356 366
  97. 97. Yap K. Y. Chui W. K. Chan A. Drug interactions. between chemotherapeutic. regimens antiepileptics Clin Ther 30 2008 8 1385 1407
  98. 98. Kristensen CA, Kristjansen PE, Hansen HH: Systemic chemotherapy of brain metastases from small-cell lung cancer: a review.J Clin Oncol 10 1992 9 1498 1502

Written By

Ryuya Yamanaka

Submitted: 11 November 2010 Published: 23 August 2011