Skip to main content

Advertisement

Log in

Current Treatment Options for Breast Cancer Brain Metastases

  • Neuro-oncology (GJ Lesser, Section Editor)
  • Published:
Current Treatment Options in Oncology Aims and scope Submit manuscript

Opinion statement

In the past, the standard of care for treatment of BM was whole brain radiation therapy (WBRT), stereotactic radiosurgery (SRS), and surgery. There has been a greater role for medical therapies in the last two decades due to the discovery of driver mutations and corresponding targeted therapies. These innovations have dramatically altered the approach to treating these patients. Some of the important mutations include epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) mutations in small cell lung cancer, human epidermal growth factor receptor (HER2) mutation in breast cancer, and BRAF mutation in melanoma. Disease-specific graded prognostic assessments have identified prognostic factors for each of the major tumor types associated with BM. These reflect the increased treatment sensitivity of these tumors to specific agents. Furthermore, there is a difference in the genetic makeup of BM compared to their primary tumor. Genomic studies of BM patients comparing somatic point mutations and copy number variations with their primary tumor have demonstrated that while both the primary tumor and BM share a number of common mutations, BM can often develop distinct mutations. Therefore, there is a need to individualize systemic therapies in BM. Several organizations including the Food and Drug Administration and the American Society of Clinical Oncology now emphasize the inclusion of BM patients in various phases of clinical drug development.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References and Recommended Reading

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. Lin, N. U., Amiri-Kordestani, L., Palmieri, D., Liewehr, D. J. & Steeg, P. S. CNS Metastases in breast cancer: old challenge, new frontiers CME Staff Planners’ disclosures acknowledgment of financial or other support. Clin Cancer Res 19, (2013).

  2. Smid M, et al. Subtypes of breast cancer show preferential site of relapse. Cancer Res. 2008;68:3108–14.

    CAS  PubMed  Google Scholar 

  3. Venur VA, Ahluwalia MS. Prognostic scores for brain metastasis patients: use in clinical practice and trial design. Chin Clin Oncol. 2015;4:18.

    PubMed  Google Scholar 

  4. Sperduto PW, Berkey B, Gaspar LE, Mehta M, Curran W. A new prognostic index and comparison to three other indices for patients with brain metastases: an analysis of 1960 patients in the RTOG Database. Int J Radiat Oncol. 2008;70:510–4.

    Google Scholar 

  5. • Sperduto PW, et al. Effect of tumor subtype on survival and the graded prognostic assessment for patients with breast cancer and brain metastases. Int J Radiat Oncol Biol Phys. 2012;82:2111–7 The authors further refined the Breast Graded Prognostic Assessment (GPA) by using multivariate Cox regression (MCR) and recursive partitioning analysis (RPA) on a multi-institutional, retrospective cohort of N = 400 breast cancer patients with newly diagnosed brain metastases to identify prognostic factors related to survival. In doing so, they identified the following: Karnofsky Performance Status (KPS) and tumor subtypes (namely HER2, ER/PR status and the interaction between these two).

    PubMed  Google Scholar 

  6. Noordijk EM, et al. The choice of treatment of single brain metastasis should be based on extracranial tumor activity and age. Int J Radiat Oncol Biol Phys. 1994;29:711–7.

    CAS  PubMed  Google Scholar 

  7. • Brown PD, et al. Effect of radiosurgery alone vs radiosurgery with whole brain radiation therapy on cognitive function in patients with 1 to 3 brain metastases. JAMA. 2016;316:401 In this randomized, multi-institutional trial conducted across 34 centers in North America, N = 213 patients with 1–3 brain metastases were randomized to two arms: SRS alone (N = 111) vs. SRS plus WBRT (N = 102). The cohort with SRS alone demonstrated less cognitive deterioration at 3 months (63.5% vs. 91.7% in the SRS plus WBRT arm; p < 0.001) with greater quality of life preserved. There was no statistically significant difference in median overall survival between the two arms (10.4 months for SRS alone vs. 7.4 months for SRS plus WBRT; hazard ratio, p = 0.92).

    PubMed  PubMed Central  Google Scholar 

  8. Deeken JF, Loscher W. The blood-brain barrier and cancer: transporters, treatment, and Trojan Horses. Clin Cancer Res. 2007;13:1663–74.

    CAS  PubMed  Google Scholar 

  9. de Vries NA, et al. P-Glycoprotein and breast cancer resistance protein: two dominant transporters working together in limiting the brain penetration of topotecan. Clin Cancer Res. 2007;13:6440–9.

    PubMed  Google Scholar 

  10. Williams NL, et al. Phase 1 study of ipilimumab combined with whole brain radiation therapy or radiosurgery for melanoma patients with brain metastases. Int J Radiat Oncol Biol Phys. 2017;99:22–30.

    CAS  PubMed  Google Scholar 

  11. Kiess AP, et al. Stereotactic radiosurgery for melanoma brain metastases in patients receiving ipilimumab: safety profile and efficacy of combined treatment. Int J Radiat Oncol Biol Phys. 2015;92:368–75.

    PubMed  PubMed Central  Google Scholar 

  12. Skrepnik T, Sundararajan S, Cui H, Stea B. Improved time to disease progression in the brain in patients with melanoma brain metastases treated with concurrent delivery of radiosurgery and ipilimumab. Oncoimmunology. 2017;6:e1283461.

    PubMed  PubMed Central  Google Scholar 

  13. Vecht CJ, et al. Treatment of single brain metastasis: radiotherapy alone or combined with neurosurgery? Ann Neurol. 1993;33:583–90.

    CAS  PubMed  Google Scholar 

  14. • Patchell RA, et al. A randomized trial of surgery in the treatment of single metastases to the brain. N Engl J Med. 1990;322:494–500 This was an early study in which the authors randomizing patients with a single brain metastasis to one of two arms: surgical resection followed by radiotherapy (N = 25) vs. radiotherapy alone (N = 23). The combination arm demonstrated favorable results across all endpoints, including lower recurrence of tumor at site of the original metastasis (20% vs. 52%, p < 0.02), longer survival (median 40 weeks vs. 15 weeks, p < 0.01) and improved functional independence (median 38 weeks vs. 8 weeks, p < 0.005).

    CAS  PubMed  Google Scholar 

  15. Mintz AH, et al. A randomized trial to assess the efficacy of surgery in addition to radiotherapy in patients with a single cerebral metastasis. Cancer. 1996;78:1470–6.

    CAS  PubMed  Google Scholar 

  16. Pollock BE, Brown PD, Foote RL, Stafford SL, Schomberg PJ. Properly selected patients with multiple brain metastases may benefit from aggressive treatment of their intracranial disease. J Neuro-Oncol. 2003;61:73–80.

    Google Scholar 

  17. • Soffietti R, et al. Diagnosis and treatment of brain metastases from solid tumors: guidelines from the European Association of Neuro-Oncology (EANO). Neuro-Oncology. 2017;19:162–74 These published guidelines from the European Association of Neuro-Oncology (EANO) represent the consensus findings of an interdisciplinary Task Force that was assembled in 2014. These evidence-based guidelines form the foundation of clinical practice for treatment of brain metastases secondary to solid tumors. The authors provide comprehensive recommendations related to imaging, neuropathology, treatment modalities, prognosis, and staging of the cancer. The full range of treatment modalities are addressed including surgery, radiation, chemotherapy, and emerging targeted therapies.

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Morikawa A, et al. Capecitabine and lapatinib uptake in surgically resected brain metastases from metastatic breast cancer patients: a prospective study. Neuro-Oncology. 2015;17:289–95.

    CAS  PubMed  Google Scholar 

  19. Lippitz B, et al. Stereotactic radiosurgery in the treatment of brain metastases: the current evidence. Cancer Treat Rev. 2014;40:48–59.

    PubMed  Google Scholar 

  20. Sperduto PW, et al. Secondary analysis of RTOG 9508, a phase 3 randomized trial of whole-brain radiation therapy versus WBRT plus stereotactic radiosurgery in patients with 1–3 brain metastases; poststratified by the Graded Prognostic Assessment (GPA). Int J Radiat Oncol. 2014;90:526–31.

    Google Scholar 

  21. Venur VA, Ahluwalia MS. Prognostic scores for brain metastasis patients: use in clinical practice and trial design. Chin Clin Oncol. 2015;4:18.

    PubMed  Google Scholar 

  22. Yamamoto M, et al. A multi-institutional prospective observational study of stereotactic radiosurgery for patients with multiple brain metastases (JLGK0901 Study Update): Irradiation-related Complications and Long-term Maintenance of Mini-Mental State Examination Scores. Int J Radiat Oncol. 2017;99:31–40.

    Google Scholar 

  23. Auchter RM, et al. A multiinstitutional outcome and prognostic factor analysis of radiosurgery for resectable single brain metastasis. Int J Radiat Oncol Biol Phys. 1996;35:27–35.

    CAS  PubMed  Google Scholar 

  24. Muacevic A, et al. Microsurgery plus whole brain irradiation versus Gamma Knife surgery alone for treatment of single metastases to the brain: a randomized controlled multicentre phase III trial. J Neuro-Oncol. 2008;87:299–307.

    Google Scholar 

  25. Suki D, et al. Comparative risk of leptomeningeal disease after resection or stereotactic radiosurgery for solid tumor metastasis to the posterior fossa. J Neurosurg. 2008;108:248–57.

    PubMed  Google Scholar 

  26. Ahn JH, et al. Risk for leptomeningeal seeding after resection for brain metastases: implication of tumor location with mode of resection. J Neurosurg. 2012;116:984–93.

    PubMed  Google Scholar 

  27. Atalar B, et al. Risk of leptomeningeal disease in patients treated with stereotactic radiosurgery targeting the postoperative resection cavity for brain metastases. Int J Radiat Oncol. 2013;87:713–8.

    Google Scholar 

  28. Johnson MD, et al. Surgical resection of brain metastases and the risk of leptomeningeal recurrence in patients treated with stereotactic radiosurgery. Int J Radiat Oncol. 2016;94:537–43.

    Google Scholar 

  29. Minniti G, et al. Health-related quality of life in elderly patients with newly diagnosed glioblastoma treated with short-course radiation therapy plus concomitant and adjuvant temozolomide. Int J Radiat Oncol. 2013;86:285–91.

    Google Scholar 

  30. Mathieu D, et al. Tumor bed radiosurgery after resection of cerebral metastases. Neurosurgery. 2008;62:817–24.

    PubMed  Google Scholar 

  31. Minniti G, et al. Single-fraction versus multifraction (3 × 9 Gy) stereotactic radiosurgery for large (&gt;2 cm) brain metastases: a comparative analysis of local control and risk of radiation-induced brain necrosis. Int J Radiat Oncol. 2016;95:1142–8.

    Google Scholar 

  32. Boothe D, et al. Bevacizumab as a treatment for radiation necrosis of brain metastases post stereotactic radiosurgery. Neuro-Oncology. 2013;15:1257–63.

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Mounsey LA, et al. Changing natural history of HER2–positive breast cancer metastatic to the brain in the era of new targeted therapies. Clin Breast Cancer. 2018;18:29–37.

    PubMed  Google Scholar 

  34. Aoyama H, et al. Neurocognitive function of patients with brain metastasis who received either whole brain radiotherapy plus stereotactic radiosurgery or radiosurgery alone. Int J Radiat Oncol. 2007;68:1388–95.

    Google Scholar 

  35. Kocher M, et al. Adjuvant whole-brain radiotherapy versus observation after radiosurgery or surgical resection of one to three cerebral metastases: results of the EORTC 22952–26,001 Study. J Clin Oncol. 2011;29:134–41.

    PubMed  Google Scholar 

  36. Brown PD, et al. NCCTG N0574 (Alliance): A phase III randomized trial of whole brain radiation therapy (WBRT) in addition to radiosurgery (SRS) in patients with 1 to 3 brain metastases. J Clin Oncol. 2015;33:LBA4–4.

  37. Brown PD, et al. Postoperative stereotactic radiosurgery compared with whole brain radiotherapy for resected metastatic brain disease (NCCTG N107C/CEC·3): a multicentre, randomised, controlled, phase 3 trial. Lancet Oncol. 2017;18:1049–60.

    PubMed  PubMed Central  Google Scholar 

  38. Brown PD, et al. Memantine for the prevention of cognitive dysfunction in patients receiving whole-brain radiotherapy: a randomized, double-blind, placebo-controlled trial. Neuro-Oncology. 2013;15:1429–37.

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Rapp SR, et al. Donepezil for irradiated brain tumor survivors: a phase III randomized placebo-controlled clinical trial. J Clin Oncol. 2015;33:1653–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Suh JH. Hippocampal-avoidance whole-brain radiation therapy: a new standard for patients with brain metastases? J Clin Oncol. 2014;32:3789–91.

    PubMed  PubMed Central  Google Scholar 

  41. Byun K-D, et al. Eribulin mesylate combined with local treatment for brain metastasis from breast cancer: two case reports. J Breast Cancer. 2016;19:214–7.

    PubMed  PubMed Central  Google Scholar 

  42. Cortés J, et al. Prolonged survival in patients with breast cancer and a history of brain metastases: results of a preplanned subgroup analysis from the randomized phase III BEACON trial. Breast Cancer Res Treat. 2017;165:329–41.

    PubMed  PubMed Central  Google Scholar 

  43. Régina A, et al. Antitumour activity of ANG1005, a conjugate between paclitaxel and the new brain delivery vector Angiopep-2. Br J Pharmacol. 2008;155:185–97.

    PubMed  PubMed Central  Google Scholar 

  44. Chamberlain M, et al. Leptomeningeal metastasis: a response assessment in neuro-oncology critical review of endpoints and response criteria of published randomized clinical trials. Neuro-Oncology. 2014;16:1176–85.

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Romond EH, et al. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med. 2005;353:1673–84.

    CAS  PubMed  Google Scholar 

  46. Leyland-Jones B. Human epidermal growth factor receptor 2–positive breast cancer and central nervous system metastases. J Clin Oncol. 2009;27:5278–86.

    PubMed  Google Scholar 

  47. Palmieri D, et al. HER-2 overexpression increases the metastatic outgrowth of breast cancer cells in the brain. Cancer Res. 2007;67:4190–8.

    CAS  PubMed  Google Scholar 

  48. Dijkers EC, et al. Biodistribution of 89Zr-trastuzumab and PET imaging of HER2-positive lesions in patients with metastatic breast cancer. Clin Pharmacol Ther. 2010;87:586–92.

    CAS  PubMed  Google Scholar 

  49. Tamura K, et al. 64Cu-DOTA-Trastuzumab PET imaging in patients with HER2-positive breast cancer. J Nucl Med. 2013;54:1869–75.

    CAS  PubMed  Google Scholar 

  50. Le Scodan R, et al. Brain metastases from breast cancer: prognostic significance of HER-2 overexpression, effect of trastuzumab and cause of death. BMC Cancer. 2011;11:395.

    PubMed  PubMed Central  Google Scholar 

  51. Park IH, et al. Trastuzumab treatment beyond brain progression in HER2-positive metastatic breast cancer. Ann Oncol. 2008;20:56–62.

    PubMed  Google Scholar 

  52. Swain SM, et al. Pertuzumab, trastuzumab, and docetaxel in HER2-positive metastatic breast cancer. N Engl J Med. 2015;372:724–34.

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Swain SM, et al. Incidence of central nervous system metastases in patients with HER2-positive metastatic breast cancer treated with pertuzumab, trastuzumab, and docetaxel: results from the randomized phase III study CLEOPATRA. Ann Oncol. 2014;25:1116–21.

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Tsukada Y, Fouad A, Pickren JW, Lane WW. Central nervous system metastasis from breast carcinoma. Autopsy Study Cancer. 1983;52:2349–54.

    CAS  PubMed  Google Scholar 

  55. de Azevedo CRAS, et al. Meningeal carcinomatosis in breast cancer: prognostic factors and outcome. J Neuro-Oncol. 2011;104:565–72.

    Google Scholar 

  56. Mir O, Ropert S, Alexandre J, Lemare F, Goldwasser F. High-dose intrathecal trastuzumab for leptomeningeal metastases secondary to HER-2 overexpressing breast cancer. Ann Oncol. 2008;19:1978–80.

    CAS  PubMed  Google Scholar 

  57. Stemmler H-J, et al. Ratio of trastuzumab levels in serum and cerebrospinal fluid is altered in HER2-positive breast cancer patients with brain metastases and impairment of blood???brain barrier. Anti-Cancer Drugs. 2007;18:23–8.

    CAS  PubMed  Google Scholar 

  58. Gulia S, Gupta S, Singh A. Intrathecal trastuzumab for leptomeningeal carcinomatosis in patients with human epidermal growth factor receptor 2 positive breast cancer. Indian J Med Paediatr Oncol. 2016;37:196–8.

    PubMed  PubMed Central  Google Scholar 

  59. Nieder C, Andratschke N, Grosu AL, Molls M. Recursive partitioning analysis (RPA) class does not predict survival in patients with four or more brain metastases. Strahlenther Onkol. 2003;179:16–20.

    PubMed  Google Scholar 

  60. LoRusso PM, Weiss D, Guardino E, Girish S, Sliwkowski MX. Trastuzumab emtansine: a unique antibody-drug conjugate in development for human epidermal growth factor receptor 2-positive cancer. Clin Cancer Res. 2011;17:6437–47.

    CAS  PubMed  Google Scholar 

  61. Verma S, et al. Trastuzumab emtansine for HER2-positive advanced breast cancer. N Engl J Med. 2012;367:1783–91.

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Krop IE, et al. Trastuzumab emtansine (T-DM1) versus lapatinib plus capecitabine in patients with HER2-positive metastatic breast cancer and central nervous system metastases: a retrospective, exploratory analysis in EMILIA. Ann Oncol. 2015;26:113–9.

    CAS  PubMed  Google Scholar 

  63. Murthy R, et al. Tucatinib with capecitabine and trastuzumab in advanced HER2-positive metastatic breast cancer with and without brain metastases: a non-randomised, open-label, phase 1b study. Lancet Oncol. 2018;19:880–8.

    CAS  PubMed  Google Scholar 

  64. Taskar KS, et al. Lapatinib distribution in HER2 overexpressing experimental brain metastases of breast cancer. Pharm Res. 2012;29:770–81.

    CAS  PubMed  Google Scholar 

  65. Polli JW, et al. The role of efflux and uptake transporters in N- -6-[5-(methyl)-2-furyl]-4-quinazolinamine (GW572016, Lapatinib) disposition and drug interactions. Drug Metab Dispos. 2008;36:695–701.

    CAS  PubMed  Google Scholar 

  66. Burris HA, et al. Phase I safety, pharmacokinetics, and clinical activity study of lapatinib (GW572016), a reversible dual inhibitor of epidermal growth factor receptor tyrosine kinases, in heavily pretreated patients with metastatic carcinomas. J Clin Oncol. 2005;23:5305–13.

    CAS  PubMed  Google Scholar 

  67. Spector NL, et al. Study of the biologic effects of lapatinib, a reversible inhibitor of ErbB1 and ErbB2 tyrosine kinases, on tumor growth and survival pathways in patients with advanced malignancies. J Clin Oncol. 2005;23:2502–12.

    CAS  PubMed  Google Scholar 

  68. •• Bachelot T, et al. Lapatinib plus capecitabine in patients with previously untreated brain metastases from HER2-positive metastatic breast cancer (LANDSCAPE): a single-group phase 2 study. Lancet Oncol. 2013;14:64–71 In this phase 2 open-label, single-arm, multi-institutional trial, HER2-positive breast cancer patients (N = 45, no prior treatment with WBRT or either study drug) with brain metastates received oral capecitabine (2000 mg/m2) daily from days 1–14 and oral lapatinib (1250 mg) daily over 21-day cycles. Twenty-nine of the 45 enrolled patients (65%) experienced an objective CNS response (95% CI 50.1–79.5%). Adverse event rates associated with the combination were acceptable with 49% of patients experiencing grade 3 or 4 events with diarrhea being the most common (20%). Results of this study laid the foundation of a follow-on phase 3 trial.

    CAS  PubMed  Google Scholar 

  69. Lin NU, et al. Multicenter phase II study of lapatinib in patients with brain metastases from HER2-positive breast cancer. Clin Cancer Res. 2009;15:1452–9.

    CAS  PubMed  Google Scholar 

  70. Lin NU, et al. Phase II trial of lapatinib for brain metastases in patients with human epidermal growth factor receptor 2-positive breast cancer. J Clin Oncol. 2008;26:1993–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  71. • Lin NU, et al. Randomized phase II study of lapatinib plus capecitabine or lapatinib plus topotecan for patients with HER2-positive breast cancer brain metastases. J Neuro-Oncol. 2011;105:613–20 This study assessed CNS activity of lapatinib (a novel EGFR/HER2 inhibitor) in N = 242 HER2+ breast cancer patients with progressive brain metastases. Only 6% of these enrolled patients met the primary outcome of CNS objective response. Response rates were more favorable in the 50 patients who enrolled in a follow-on lapatinib plus capecitabine extension arm with 20% meeting the CNS objective response criteria.

  72. Zhao X, et al. Neratinib reverses ATP-binding cassette B1-mediated chemotherapeutic drug resistance in vitro, in vivo, and ex vivo. Mol Pharmacol. 2012;82:47–58.

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Bose R, et al. Activating HER2 mutations in HER2 gene amplification negative breast cancer. Cancer Discov. 2013;3:224–37.

    CAS  PubMed  Google Scholar 

  74. Canonici A, et al. Neratinib overcomes trastuzumab resistance in HER2 amplified breast cancer. Oncotarget. 2013;4:1592–605.

    PubMed  PubMed Central  Google Scholar 

  75. Freedman RA, et al. Translational Breast Cancer Research Consortium (TBCRC) 022: a phase II trial of neratinib for patients with human epidermal growth factor receptor 2-positive breast cancer and brain metastases. J Clin Oncol. 2016;34:945–52.

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Freedman RA, et al. TBCRC 022: Phase II trial of neratinib + capecitabine for patients (Pts) with human epidermal growth factor receptor 2 (HER2+) breast cancer brain metastases (BCBM). J Clin Oncol. 2017;35:1005–5.

  77. Saura C, et al. Safety and efficacy of neratinib in combination with capecitabine in patients with metastatic human epidermal growth factor receptor 2–positive breast cancer. J Clin Oncol. 2014;32:3626–33.

    CAS  PubMed  Google Scholar 

  78. Hamilton, E., Borges, V., Conlin, A., Walker, L. & Moulder, S. Abstract P4-21-01: efficacy results of a phase 1b study of ONT-380, an oral HER2-specific inhibitor, in combination with capecitabine (C) and trastuzumab (T) in HER2+ metastatic breast cancer (MBC), including patients (pts) with brain metastases (mets). Cancer Res. 77, P4–21-01-P4–21-01 (2017).

  79. Borges VF, et al. Tucatinib combined with ado-trastuzumab emtansine in advanced ERBB2/HER2-positive metastatic breast cancer. JAMA Oncol. 2018;4:1214.

    PubMed  Google Scholar 

  80. Lee JJ, Loh K, Yap Y-S. PI3K/Akt/mTOR inhibitors in breast cancer. Cancer Biol Med. 2015;12:342–54.

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Baselga J, et al. Everolimus in postmenopausal hormone-receptor–positive advanced breast cancer. N Engl J Med. 2012;366:520–9.

    CAS  PubMed  Google Scholar 

  82. André F, et al. Everolimus for women with trastuzumab-resistant, HER2-positive, advanced breast cancer (BOLERO-3): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet Oncol. 2014;15:580–91.

    PubMed  Google Scholar 

  83. Cox MC, Dan TD, Swain SM. Emerging drugs to replace current leaders in first-line therapy for breast cancer. Expert Opin Emerg Drugs. 2006;11:489–501.

    CAS  PubMed  Google Scholar 

  84. Miller K, et al. Paclitaxel plus bevacizumab versus paclitaxel alone for metastatic breast cancer. N Engl J Med. 2007;357:2666–76.

    CAS  PubMed  Google Scholar 

  85. Miles DW, et al. First-line bevacizumab in combination with chemotherapy for HER2-negative metastatic breast cancer: pooled and subgroup analyses of data from 2447 patients. Ann Oncol Off J Eur Soc Med Oncol. 2013;24:2773–80.

    CAS  Google Scholar 

  86. Lu Y-S, et al. Bevacizumab preconditioning followed by etoposide and cisplatin is highly effective in treating brain metastases of breast cancer progressing from whole-brain radiotherapy. Clin Cancer Res. 2015;21:1851–8.

    CAS  PubMed  Google Scholar 

  87. Choi YJ, Anders L. Signaling through cyclin D-dependent kinases. Oncogene. 2014;33:1890–903.

    CAS  PubMed  Google Scholar 

  88. Hamilton E, Infante JR. Targeting CDK4/6 in patients with cancer. Cancer Treat Rev. 2016;45:129–38.

    CAS  PubMed  Google Scholar 

  89. Turner NC, et al. Palbociclib in hormone-receptor–positive advanced breast cancer. N Engl J Med. 2015;373:209–19.

    CAS  PubMed  Google Scholar 

  90. Stemmler H-J, Heinemann V. Central nervous system metastases in HER-2-overexpressing metastatic breast cancer: a treatment challenge. Oncologist. 2008;13:739–50.

    PubMed  Google Scholar 

  91. Cortés J, et al. Afatinib alone or afatinib plus vinorelbine versus investigator’s choice of treatment for HER2-positive breast cancer with progressive brain metastases after trastuzumab, lapatinib, or both (LUX-Breast 3): a randomised, open-label, multicentre, phase 2 trial. Lancet Oncol. 2015;16:1700–10.

    PubMed  Google Scholar 

  92. Bachelot T, et al. Lapatinib plus capecitabine in patients with previously untreated brain metastases from HER2-positive metastatic breast cancer (LANDSCAPE): a single-group phase 2 study. Lancet Oncol. 2013;14:64–71.

    CAS  PubMed  Google Scholar 

  93. Ribas, A. & Wolchok, J. D. Cancer immunotherapy using checkpoint blockade. Science (80-.). 359, 1350–1355 (2018).

  94. Blank CU, Enk A. Therapeutic use of anti-CTLA-4 antibodies. Int Immunol. 2015;27:3–10.

    CAS  PubMed  Google Scholar 

  95. Lin NU, et al. Phase II trial of lapatinib for brain metastases in patients with human epidermal growth factor receptor 2–positive breast cancer. J Clin Oncol. 2008;26:1993–9.

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Manmeet S. Ahluwalia MD.

Ethics declarations

Conflict of Interest

Arrvind Raghunath declares that he has no conflict of interest. Kunal Desai declares that he has no conflict of interest. Manmeet S. Ahluwalia has received research funding from Incyte, Bristol-Myers Squibb, AstraZeneca, TRACON, Novartis, Novocure, AbbVie, Pharmacyclics, Merck, and Bayer; has received compensation from Incyte, Bristol-Myers Squibb, AstraZeneca, Novocure, Monteris Medical, Caris Life Sciences, MR Solutions, AbbVie, CBT Pharmaceuticals, Flatiron Health, Varian Medical Systems, and MimiVax for service as a consultant; has received compensation from VBI Vaccines for participating on a data and safety monitoring board; and has stock options in MimiVax and Doctible.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

This article is part of the Topical Collection on Neuro-oncology

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Raghunath, A., Desai, K. & Ahluwalia, M.S. Current Treatment Options for Breast Cancer Brain Metastases. Curr. Treat. Options in Oncol. 20, 19 (2019). https://doi.org/10.1007/s11864-019-0618-5

Download citation

  • Published:

  • DOI: https://doi.org/10.1007/s11864-019-0618-5

Keywords

Navigation