Skip to main content

Advertisement

Log in

Molecular Subtyping of Brain Metastases and Implications for Therapy

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

Opinion statement

Molecular subtyping of tumors and treatment with specifically targeted therapy is a rapidly developing trend in oncology. Genetic and protein biomarkers impact biological behavior, patient prognosis, and inform treatment options. Select examples include EGFR mutations in primary non-small cell lung cancers, Her2 overexpression in breast cancer, and BRAF mutations in melanoma. Systemic benefit is emphasized in targeted therapies; yet lung cancer, breast cancer, and melanoma comprise the most common diagnoses in patients with brain metastases making the effectiveness of targeted therapies in the treatment and/or prevention of brain metastases relevant.Emerging evidence suggests efficacy for targeted therapy in the setting of brain metastases. Randomized, phase III clinical trials indicate targeted HER2 treatment with lapatinib and capecitabine in brain metastases from breast cancer increases the time to progression and decreases the frequency of CNS involvement at progression. Phase II trials and retrospective reviews for gefitinib and erlotinib demonstrate these agents may have a role in both the chemoprevention of brain metastases and, in combination with WBRT, treatment for non-small cell lung cancer (NSCLC) brain metastases. Dabrafenib and other BRAF inhibitors have demonstrated improved survival in patients with brain metastases from melanoma in a recent phase II clinical trial. Further data that support the use of these agents are the subject of several active clinical trials. Challenges and future directions for targeted therapies in brain metastases include both better characterization and drug design with respect to central nervous system distribution. Limited published data demonstrate suboptimal CNS distribution of currently available targeted chemotherapeutic agents. Increasing systemic dosing, alternate delivery methods, and new compounds with improved CNS distribution are being pursued. Additionally, eventual resistance to targeted therapies poses a challenge; however, research is showing resistance mutations are conserved and relatively predictable creating opportunities for second-line therapies with additional targeted drugs. Newer targeted therapies represent an additional chemotherapeutic option for the treatment and/or prevention of brain metastases in patients with an appropriate molecular profile.

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. Patchell RA. The management of brain metastases. Cancer Treat Rev. 2003;29(6):533–40.

    Article  PubMed  Google Scholar 

  2. Mehta M, Vogelbaum M, Chang S, Patel N. Neoplasms of the central nervous system. In: DeVita Jr VT, Lawrence TSRS, editors. Cancer: Principles and practice of oncology. 9th ed. Philadelphia: Lippincott Williams & Wilkins; 2011. p. 1700–49.

    Google Scholar 

  3. Tsao MN, Lloyd N, Wong RKS, et al. Whole brain radiotherapy for the treatment of newly diagnosed multiple brain metastases. Cochrane Database Syst Rev (Online). 2012;4:CD003869.

    Google Scholar 

  4. Gaspar L, Scott C, Rotman M, et al. Recursive partitioning analysis (RPA) of prognostic factors in three Radiation Therapy Oncology Group (RTOG) brain metastases trials. Int J Radiat Oncol Biol Phys. 1997;37(4):745–51. A widely used clinical tool in estimating prognosis and guiding treatment decision making for patients with brain metastases.

    Article  PubMed  CAS  Google Scholar 

  5. 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):419–25. An excellent resource containing easy-to-use worksheet for the most common brain metastases and has customized GPA based on tumor histology. Also contains comprehensive, multi-institutional survival data for patients with brain metastases.

    Article  PubMed  Google Scholar 

  6. Sperduto PW, Kased N, Roberge D, 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(5):2111–7.

    Article  PubMed  Google Scholar 

  7. Posner JB. Management of central nervous system metastases. Semin Oncol. 1977;4(1):81–91.

    PubMed  CAS  Google Scholar 

  8. Zimm S, Wampler GL, Stablein D, et al. Intracerebral metastases in solid-tumor patients: natural history and results of treatment. Cancer. 1981;48(2):384–94.

    Article  PubMed  CAS  Google Scholar 

  9. Patchell RA, Tibbs PA, Regine WF, et al. Postoperative radiotherapy in the treatment of single metastases to the brain: a randomized trial. JAMA. 1998;280(17):1485–9.

    Article  PubMed  CAS  Google Scholar 

  10. Chao JH, Phillips R, Nickson JJ. Roentgen-ray therapy of cerebral metastases. Cancer. 1954;7(4):682–9.

    Article  PubMed  CAS  Google Scholar 

  11. Borgelt B, Gelber R, Kramer S, et al. The palliation of brain metastases: final results of the first two studies by the Radiation Therapy Oncology Group. Int J Radiat Oncol Biol Phys. 1980;6(1):1–9.

    Article  PubMed  CAS  Google Scholar 

  12. Andrews DW, Scott CB, Sperduto PW, et al. Whole brain radiation therapy with or without stereotactic radiosurgery boost for patients with one to three brain metastases: phase III results of the RTOG 9508 randomised trial. Lancet. 2004;363(9422):1665–72.

    Article  PubMed  Google Scholar 

  13. Patil CG, Pricola K, Sarmiento JM, et al. Whole brain radiation therapy (WBRT) alone versus WBRT and radiosurgery for the treatment of brain metastases. Cochrane Database Syst Rev (Online). 2012;9:CD006121.

    Google Scholar 

  14. Robbins JR, Ryu S, Kalkanis S, et al. Radiosurgery to the surgical cavity as adjuvant therapy for resected brain metastasis. Neurosurgery. 2012;71(5):937–43. Summarized the current studies investigating this commonly used treatment strategy and reports local and distant control rates.

    Article  PubMed  Google Scholar 

  15. Kocher M, Soffietti R, Abacioglu U, et al. Adjuvant whole-brain radiotherapy versus observation after radiosurgery or surgical resection of one to three cerebral metastases: results of the EORTC 22952-26001 study. J Clin Oncol. 2011;29(2):134–41. Phase III trial demonstrating no difference in survival when WBRT is omitted after SRS. However, local failure and distant failure are more common highlighting the important of routine surveillence imaginag.

    Article  PubMed  Google Scholar 

  16. Aoyama H, Shirato H, Tago M, et al. Stereotactic radiosurgery plus whole-brain radiation therapy vs stereotactic radiosurgery alone for treatment of brain metastases: a randomized controlled trial. JAMA. 2006;295(21):2483–91.

    Article  PubMed  CAS  Google Scholar 

  17. Chang EL, Wefel JS, Hess KR, et al. Neurocognition in patients with brain metastases treated with radiosurgery or radiosurgery plus whole-brain irradiation: a randomised controlled trial. Lancet Oncol. 2009;10(11):1037–44.

    Article  PubMed  Google Scholar 

  18. Patchell RA, Tibbs PA, Walsh JW, et al. A randomized trial of surgery in the treatment of single metastases to the brain. N Engl J Med. 1990;322(8):494–500.

    Article  PubMed  CAS  Google Scholar 

  19. Vecht CJ, Haaxma-Reiche H, Noordijk EM, et al. Treatment of single brain metastasis: radiotherapy alone or combined with neurosurgery? Ann Neurol. 1993;33(6):583–90.

    Article  PubMed  CAS  Google Scholar 

  20. Mintz AH, Kestle J, Rathbone MP, 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(7):1470–6.

    Article  PubMed  CAS  Google Scholar 

  21. Bindal RK, Sawaya R, Leavens ME, Lee JJ. Surgical treatment of multiple brain metastases. J Neurosurg. 1993;79(2):210–6.

    Article  PubMed  CAS  Google Scholar 

  22. Stark AM, Tscheslog H, Buhl R, et al. Surgical treatment for brain metastases: prognostic factors and survival in 177 patients. Neurosurg Rev. 2005;28(2):115–9.

    Article  PubMed  Google Scholar 

  23. Paek SH, Audu PB, Sperling MR, et al. Reevaluation of surgery for the treatment of brain metastases: review of 208 patients with single or multiple brain metastases treated at one institution with modern neurosurgical techniques. Neurosurgery. 2005;56(5):1021–34. discussion 1021–34.

    PubMed  Google Scholar 

  24. Slamon DJ, Clark GM, Wong SG, et al. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science (New York). 1987;235(4785):177–82.

    Article  CAS  Google Scholar 

  25. Pestalozzi BC, Zahrieh D, Price KN, et al. Identifying breast cancer patients at risk for Central Nervous System (CNS) metastases in trials of the International Breast Cancer Study Group (IBCSG). Ann Oncol. 2006;17(6):935–44.

    Article  PubMed  CAS  Google Scholar 

  26. Yau T, Swanton C, Chua S, et al. Incidence, pattern and timing of brain metastases among patients with advanced breast cancer treated with trastuzumab. Acta Oncol (Stockholm, Sweden). 2006;45(2):196–201.

    Article  CAS  Google Scholar 

  27. Stemmler HJ, Kahlert S, Siekiera W, et al. Characteristics of patients with brain metastases receiving trastuzumab for HER2 overexpressing metastatic breast cancer. Breast (Edin). 2006;15(2):219–25.

    Article  CAS  Google Scholar 

  28. Shmueli E, Wigler N, Inbar M. Central nervous system progression among patients with metastatic breast cancer responding to trastuzumab treatment. Eur J Cancer (Oxford 1990). 2004;40(3):379–82.

    Article  CAS  Google Scholar 

  29. Clayton AJ, Danson S, Jolly S, et al. Incidence of cerebral metastases in patients treated with trastuzumab for metastatic breast cancer. Br J Cancer. 2004;91(4):639–43.

    PubMed  CAS  Google Scholar 

  30. Bendell JC, Domchek SM, Burstein HJ, et al. Central nervous system metastases in women who receive trastuzumab-based therapy for metastatic breast carcinoma. Cancer. 2003;97(12):2972–7.

    Article  PubMed  Google Scholar 

  31. Pinder MC, Chang H, Broglio LB, et al. Trastuzumab treatment and the risk of central nervous system (CNS) metastases [abstract]. In: Journal of Clinical Oncology, ASCO Annual Meeting Proceedings; 2007 June 2; Chicago, Illinois (IL): ASCO; 2007; 25(18S):1018.

  32. Leyland-Jones B. Human epidermal growth factor receptor 2-positive breast cancer and central nervous system metastases. J Clin Oncol. 2009;27(31):5278–86.

    Article  PubMed  Google Scholar 

  33. Lower EE, Drosick DR, Blau R, et al. Increased rate of brain metastasis with trastuzumab therapy not associated with impaired survival. Clin Breast Cancer. 2003;4(2):114–9.

    Article  PubMed  CAS  Google Scholar 

  34. Bria E, Cuppone F, Fornier M, et al. Cardiotoxicity and incidence of brain metastases after adjuvant trastuzumab for early breast cancer: the dark side of the moon? A meta-analysis of the randomized trials. Breast Cancer Res Treat. 2008;109(2):231–9.

    Article  PubMed  CAS  Google Scholar 

  35. Fokstuen T, Wilking N, Rutqvist LE, et al. Radiation therapy in the management of brain metastases from breast cancer. Breast Cancer Res Treat. 2000;62(3):211–6.

    Article  PubMed  CAS  Google Scholar 

  36. Berghoff AS, Bago-Horvath Z, Dubsky P, et al. Impact of HER-2-targeted therapy on overall survival in patients with HER-2 positive metastatic breast cancer. Breast J. 2013;19(2):149–55.

    Article  PubMed  CAS  Google Scholar 

  37. Pestalozzi BC, Brignoli S. Trastuzumab in CSF. J Clin Oncol. 2000;18(11):2349–51.

    PubMed  CAS  Google Scholar 

  38. Baselga J, Cortés J, Kim S-B, et al. Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancer. N Engl J Med. 2012;366(2):109–19.

    Article  PubMed  CAS  Google Scholar 

  39. Verma S, Miles D, Gianni L, et al. Trastuzumab emtansine for HER2-positive advanced breast cancer. N Engl J Med. 2012;367(19):1783–91.

    Article  PubMed  CAS  Google Scholar 

  40. Lin NU, Diéras V, Paul D, et al. Multicenter phase II study of lapatinib in patients with brain metastases from HER2-positive breast cancer. Clin Cancer Res. 2009;15(4):1452–9.

    Article  PubMed  CAS  Google Scholar 

  41. Lin NU, Carey LA, Liu MC, 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(12):1993–9.

    Article  PubMed  CAS  Google Scholar 

  42. Bachelot T, Romieu G, Campone M, et al. LANDSCAPE: An FNCLCC phase II study with lapatinib (L) and capecitabine (C) in patients with brain metastases (BM) from HER2-positive (+) metastatic breast cancer (MBC) before whole-brain radiotherapy (WBR) [abstract]. In: Journal of Clinical Oncology, ASCO Annual Meeting Proceedings; 2011 June 9; Chicago, Illinois (IL): ASCO; 2011; 29(15S): 509.

  43. Cameron D, Casey M, Press M, et al. A phase III randomized comparison of lapatinib plus capecitabine versus capecitabine alone in women with advanced breast cancer that has progressed on trastuzumab: updated efficacy and biomarker analyses. Breast Cancer Res Treat. 2008;112(3):533–43.

    Article  PubMed  CAS  Google Scholar 

  44. Burstein HJ, Sun Y, Dirix LY, et al. Neratinib, an irreversible ErbB receptor tyrosine kinase inhibitor, in patients with advanced ErbB2-positive breast cancer. J Clin Oncol. 2010;28(8):1301–7.

    Article  PubMed  CAS  Google Scholar 

  45. Patel AG, De Lorenzo SB, Flatten KS, et al. Failure of iniparib to inhibit poly(ADP-Ribose) polymerase in vitro. Clin Cancer Res. 2012;18(6):1655–62.

    Article  PubMed  CAS  Google Scholar 

  46. Moulder S, Mita M, Rocha C HL. A phase 1b study to assess the safety and tolerability of iniparib (BSI-201) in combination with irinotecan for the treatment of patients with Metastatic Breast Cancer (MBC) [abstract]. In: Proceedings of the 33rd Annual San Antonie Breast Cancer Symposium; 2010 Dec 12; San Antonio, Texas (TX): Proc SABCS; 2010: P-15-01.

  47. Mamon HJ, Yeap BY, Jänne PA, et al. High risk of brain metastases in surgically staged IIIA non-small-cell lung cancer patients treated with surgery, chemotherapy, and radiation. J Clin Oncol. 2005;23(7):1530–7.

    Article  PubMed  Google Scholar 

  48. Ricciardi S, De Marinis F. Multimodality management of non-small cell lung cancer patients with brain metastases. Curr Opin Oncol. 2010;22(2):86–93.

    Article  PubMed  CAS  Google Scholar 

  49. Wu C, Li YL, Wang ZM, et al. Gefitinib as palliative therapy for lung adenocarcinoma metastatic to the brain. Lung Cancer (Amsterdam, Netherlands). 2007;57(3):359–64.

    Article  Google Scholar 

  50. Chen AM, Jahan TM, Jablons DM, et al. Risk of cerebral metastases and neurological death after pathological complete response to neoadjuvant therapy for locally advanced nonsmall-cell lung cancer: clinical implications for the subsequent management of the brain. Cancer. 2007;109(8):1668–75.

    Article  PubMed  Google Scholar 

  51. Paez JG, Jänne PA, Lee JC, et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science (New York). 2004;304(5676):1497–500.

    Article  CAS  Google Scholar 

  52. Pao W, Miller V, Zakowski M, et al. EGF receptor gene mutations are common in lung cancers from “never smokers” and are associated with sensitivity of tumors to gefitinib and erlotinib. Proc Natl Acad Sci U S A. 2004;101(36):13306–11.

    Article  PubMed  CAS  Google Scholar 

  53. Varella-Garcia M, Berry L, Su P, et al. ALK and MET genes in advanced lung adenocarcinomas: The Lung Cancer Mutation Consortium experience [abstract]. In: Journal of Clinical Oncology, ASCO Annual Meeting Proceedings; 2012 May 20; Chicago, Illinois (IL): ASCO; 2012; 30(15S): 7589. Large, retrospective analysis of more than 900 patients for ALK gene rearrangements identifies the frequency and clinical correlates with this mutation.

  54. Wang M, Jing ZMC, Minjiang C. Cerebral penetration of gefitinib in patients with lung adenocarcinoma. [abstract]. In: Journal of Clinical Oncology, ASCO Annual Meeting Proceedings; 2011 June 9; Chicago, Illinois (IL): ASCO; 2011; 29(15S): 7608.

  55. Togashi Y, Masago K, Masuda S, et al. Cerebrospinal fluid concentration of gefitinib and erlotinib in patients with non-small cell lung cancer. Cancer Chemother Pharmacol. 2012;70(3):399–405.

    Article  PubMed  CAS  Google Scholar 

  56. Lee DH, Han J-Y, Lee HG, et al. Gefitinib as a first-line therapy of advanced or metastatic adenocarcinoma of the lung in never-smokers. Clin Cancer Res. 2005;11(8):3032–7.

    Article  PubMed  CAS  Google Scholar 

  57. Kim J-E, Lee DH, Choi Y, et al. Epidermal growth factor receptor tyrosine kinase inhibitors as a first-line therapy for never-smokers with adenocarcinoma of the lung having asymptomatic synchronous brain metastasis. Lung Cancer (Amsterdam, Netherlands). 2009;65(3):351–4.

    Article  Google Scholar 

  58. Heon S, Yeap BY, Lindeman NI, et al. The impact of initial gefitinib or erlotinib versus chemotherapy on central nervous system progression in advanced non-small cell lung cancer with EGFR mutations. Clin Cancer Res. 2012;18(16):4406–14. Retrospective study of 154 patients investigating up front treatment with tyrosine kinase inhibitors. One of the only studies using patients screened for EGFR mutations. At 1 year, the rate of CNS progression was 6% in the tyrosine kinase group and 19% in the chemotherapy group suggesting upfront treatment with tyrosine kinase inhibitors may prevent or delay the development of brain metastases.

    Article  PubMed  CAS  Google Scholar 

  59. Ceresoli GL, Cappuzzo F, Gregorc V, et al. Gefitinib in patients with brain metastases from non-small-cell lung cancer: a prospective trial. Ann Oncol. 2004;15(7):1042–7.

    Article  PubMed  CAS  Google Scholar 

  60. Welsh JW, Komaki R, Amini A, et al. Phase II trial of erlotinib plus concurrent whole-brain radiation therapy for patients with brain metastases from non-small-cell lung cancer. J Clin Oncol. 2013;31(7):895–902.

    Article  PubMed  CAS  Google Scholar 

  61. Costa DB, Kobayashi S, Pandya SS, et al. CSF concentration of the anaplastic lymphoma kinase inhibitor crizotinib. J Clin Oncol. 2011;29(15):e443–5.

    Article  PubMed  Google Scholar 

  62. Falk AT, Poudenx M, Otto J, et al. Adenocarcinoma of the lung with miliary brain and pulmonary metastases with echinoderm microtubule-associated protein like 4-anaplastic lymphoma kinase translocation treated with crizotinib: a case report. Lung Cancer (Amsterdam, Netherlands). 2012;78(3):282–4.

    Article  Google Scholar 

  63. Mehra R, Camidge D, Sharma S, et al. First-in-human phase I study of the ALK inhibitor LDK378 in advanced solid tumors. [abstract]. In: Journal of Clinical Oncology, ASCO Annual Meeting Proceedings; 2012 May 20; Chicago, Illinois (IL): ASCO; 2012; 30(15S): 3007.

  64. De la Monte SM, Moore GW, Hutchins GM. Patterned distribution of metastases from malignant melanoma in humans. Cancer Res. 1983;43(7):3427–33.

    PubMed  Google Scholar 

  65. Davies MA, Liu P, McIntyre S, et al. Prognostic factors for survival in melanoma patients with brain metastases. Cancer. 2011;117(8):1687–96.

    Article  PubMed  Google Scholar 

  66. Margolin K, Ernstoff MS, Hamid O, et al. Ipilimumab in patients with melanoma and brain metastases: an open-label, phase 2 trial. Lancet Oncol. 2012;13(5):459–65.

    Article  PubMed  CAS  Google Scholar 

  67. Flaherty KT, Puzanov I, Kim KB, et al. Inhibition of mutated, activated BRAF in metastatic melanoma. N Engl J Med. 2010;363(9):809–19. A 55 patient, phase I study represting the early data showing the majority of patient with the V600E BRAF mutation show at least a partial tumor response to PLX4032. The median PFS was 7 months.

    Article  PubMed  CAS  Google Scholar 

  68. Capper D, Preusser M, Habel A, et al. Assessment of BRAF V600E mutation status by immunohistochemistry with a mutation-specific monoclonal antibody. Acta Neuropathol. 2011;122(1):11–9.

    Article  PubMed  CAS  Google Scholar 

  69. Capper D, Berghoff AS, Magerle M, et al. Immunohistochemical testing of BRAF V600E status in 1,120 tumor tissue samples of patients with brain metastases. Acta Neuropathol. 2012;123(2):223–33.

    Article  PubMed  CAS  Google Scholar 

  70. Rochet NM, Kottschade LA, Markovic SN. Vemurafenib for melanoma metastases to the brain. N Engl J Med. 2011;365(25):2439–41.

    Article  PubMed  CAS  Google Scholar 

  71. Rochet NM, Dronca RS, Kottschade LA, et al. Melanoma brain metastases and vemurafenib: need for further investigation. Mayo Clin Proc Mayo Clin. 2012;87(10):976–81.

    Article  CAS  Google Scholar 

  72. Long GV, Kefford R, Carr P, et al. Phase 1/2 study of GSK2118436, a selective inhibitor of v600 mutant BRAF kinase: evidence of activity in melanoma brain metastases (Mets) [abstract]. Presented at the 25th European Society for Medical Oncology ESMO Congress; 2010 October 10; Milan, Italy: ESMO; 2010; 21(8): LBA27.

  73. Long GV, Trefzer U, Davies MA, et al. Dabrafenib in patients with Val600Glu or Val600Lys BRAF-mutant melanoma metastatic to the brain (BREAK-MB): a multicentre, open-label, phase 2 trial. Lancet Oncol. 2012;13(11):1087–95. First multi-center, phase II trial completed investigating BRAF inhibitor specifically with mets to the brian as many of these patients were excluded from the earlier PLX4032 trials. Trial enrolled 172 patients with brain mets and proven BRAF mutation. Response rates varied between 30.8% and 39.2% in patient who received prior CNS therapy vs. those who did not. The toxicity profile was acceptable with pyrexia, intracranial hemorrhage, and squamous cell carcinomas being the most common serious side effects.

    Article  PubMed  CAS  Google Scholar 

Download references

Conflicts of Interest

Glenn J. Lesser is a consultant to Genentech and received honoraria from Merck.

Jaclyn J. Renfrow declares that she has no conflict of interest.

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.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Glenn J. Lesser MD, FACP.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Renfrow, J.J., Lesser, G.J. Molecular Subtyping of Brain Metastases and Implications for Therapy. Curr. Treat. Options in Oncol. 14, 514–527 (2013). https://doi.org/10.1007/s11864-013-0248-2

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11864-013-0248-2

Keywords

Navigation