Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Oct 1.
Published in final edited form as: Crit Rev Oncol Hematol. 2019 Jul 22;142:44–50. doi: 10.1016/j.critrevonc.2019.07.012

Systemic therapy for brain metastases

Jonathan W Rick 1, Maryam Shahin 1, Ankush Chandra 1, Cecilia Dalle Ore 1, John K Yue 1, Alan Nguyen 1, Garima Yagnik 1, Soumya Sagar 1, Saman Arfaie 1, Manish K Aghi 1,*
PMCID: PMC6746616  NIHMSID: NIHMS1044176  PMID: 31357143

Abstract

Metastases from cells outside of the central nervous system are the most common cancer found in the brain and are commonly associated with poor prognosis. Although cancer treatment is improving overall, central nervous system metastases are becoming more prevalent and require finesse to properly treat. Physicians must consider the biology of the primary tumor and the complex neurological environment that the metastasis resides in. This can be further complicated by the fact that the practice of cancer management is constantly evolving and therapy that works outside of the blood-brain barrier may not be effective inside of it. Therefore, this review seeks to update the reader on recent advancements made on the three most common sources of brain metastases: lung cancer, breast cancer, and melanoma. Each of these malignancies has been the subject of intriguing and novel avenues of therapy which are reviewed here.

Keywords: Systemic therapy, Metastasis, Lung cancer, Breast cancer, Melanoma

1. Introduction

The sine qua non of cancer is the potential to spread beyond its source. This process of metastasis allows tumors to colonize diverse tissues, well beyond what could be achieved by local invasion. Although metastases can be found in any organ, they are particularly worrisome in the central nervous system (CNS) due to their propensity to cause neurological deficits (Davey, 2002; Nieder et al., 2018). Overall, tumors from foreign tissues, outside of the CNS. are much more common than primary brain cancer and only one of ten cancers found in the CNS originates there (Lassman and DeAngelis, 2003). Additionally, advancement in cancer therapy has resulted in improved survival from many malignancies and therefore more time for cancer to spread to the CNS (Davey, 2002). There has been a concordant rise in the prevalence of CNS metastases meaning that these tumors are of significant and growing concern (Nayak et al., 2012). Many different cancers can give rise to a CNS metastasis. Lung (48%) and breast (15%) cancer, along with melanoma (10%), are the three most common sources of brain metastases. Overall, 10–30% of patients with lung cancer will experience a brain metastasis, compared to 10–15% of breast cancer and up to 60% of stage 4 melanoma (Sandru et al., 2014; Leone and Leone, 2015; Niemiec et al., 2011). The incidence of brain metastases by subtype is a product of both the primary cancer’s prevalence and its propensity to metastasize to the CNS.

The challenge in treating metastases arises from their marked heterogeneity of origin and therapeutic response (Nayak et al., 2012). Treating CNS metastases requires an adroit sense of oncology and an appreciation for the neurological complications that cancer, or therapy, can induce. The goal of this article is to review the current state of systemic therapy for CNS metastases, with a special emphasis on the brain. We place a particular importance on new treatments with the goal of providing a broad update on treating the three most common brain metastases.

2. Drug delivery in the context of the blood-brain barrier

Treatment of primary brain cancer, namely glioblastoma, has been hampered by the protection afforded by the semi-permeable blood-brain barrier (BBB) (Stupp et al., 2005). This barrier is composed of numerous endothelial cells and their dense network of tight junctions (Stewart, 1994). Thus, the BBB provides much more selectivity than is typically afforded by vasculature alone, and it is thought to impede 100% of large molecule and 98% of small molecule drugs (Zhang et al., 1992).

Curiously, the BBB, which acts as a major obstacle in primary brain cancer treatment, may be a relatively minor impedance in treating some metastases (Davey, 2002; Donelli et al., 1992; Lesser, 1996). Unlike primary brain tumors, many metastases appear to respond, at least partially, to drugs that have activity against the primary tumor (Lassman and DeAngelis, 2003; Lesser, 1996; Chamberlain et al., 2017). Studies aimed at teasing apart this paradox have identified that, in the process of spreading and invading the CNS, metastases disrupt the BBB and make it permeable to many systemic agents (Lesser, 1996). This gives neuro-oncologists the opportunity to apply diverse medications against brain cancer. However, disruption of the BBB is not ubiquitous across all cancers and systemic therapy plays various roles that largely depended on the primary (Chamberlain et al., 2017; Whitsett et al., 2013). Like all systemic therapy design, the identity and context of the cancer is important. As a general concept, CNS metastases appear to exhibit similar chemosensitivies as their primary tumor (Chamberlain et al., 2017; Whitsett et al., 2013). Metastases are treated based on subtype, which informs the remaining structure of this review.

3. Specific considerations for therapies by tumor type

3.1. Lung Cancer

Lung cancer is the leading cause of cancer death in the United States and the most common source of brain metastases (Barnholtz-Sloan et al., 2004; Herbst et al., 2008). These malignancies are broadly categorized into small cell lung carcinoma (SCLC), which originate from neuroendocrine cells, and non-small cell lung carcinoma (NSCLC), which includes large cell carcinoma, squamous cell carcinoma, and adenocarcinoma (Herbst et al., 2008). This division separates out the more chemosensitive SSLC from the typically more chemoresistant NSCLC – attributes that tend to carry over to metastases.

There have been a number of studies that have identified objective responses of SCLC metastases in the CNS to systemically administered chemotherapy (Chen et al., 2008; Postmus and Smit, 1999). Most notably are Twelves et al. who reported a decrease in size in 9/14 (64%) patients that were treated with a combination of cyclophosphamide, vincristine and etoposide (Twelves et al., 1990). The findings on imaging were corroborated by a concordant reduction in neurological symptoms (Twelves et al., 1990). Kristjansen et al. had similar results with a trial of multidrug chemotherapy and reported a response in 11/13 patients and a median response time of 135 days (Kristjansen et al., 1993). Both of these studies found that CNS metastases have modestly reduced response rates when compared to extra-cranial metastases. This suggests that the BBB will at least partially limit chemotherapy diffusion, or else that cells capable of metastasizing through the BBB are more resistant to typical agents (Twelves et al., 1990; Kristjansen et al., 1993). Whole brain radiotherapy (WBRT) is the most common treatment modality for SCLC brain metastases and there has been persistent interest in combining chemotherapy with radiation. However, a meta-analysis of 3 randomized control trials (RCTs) (n = 192) found no differences in overall survival (OS) in patients treated with combined WBRT and various chemotherapeutics(Reveiz et al., 2012). SCLC, like other forms of lung cancer, carries a poor prognosis independent of dissemination to the CNS. It appears that chemotherapy is able to prolong PFS in patients with intracranial metastases from SCLC, but this does not translate to improved OS (Reveiz et al., 2012).

NSCLC is typically less responsive to chemotherapy than SCLC, and this principle appears to apply to metastases as well (Herbst et al., 2008; Dempke et al., 2015). A combination of etoposide and cisplatin affected a response rate of 30% in brain metastases for previously untreated patients, which is markedly less than what is seen in SCLC (Twelves et al., 1990). Despite numerous trials using other cytotoxic drug combinations, no systemic regimen has been able to rival radiotherapy alone (Twelves et al., 1990).

However, over the past 15 years, there has been a rise in the use of small molecule inhibitors for NSCLC. The first example of these in NSCLC are the epidermal growth factor receptor tyrosine kinase (EGFR-TKI) inhibitors, erlotinib and gefitinib, which have effects on EGFR-mutated cancers (Dempke et al., 2015). These agents have been shown to produce great responses in intracranial tumors (60–80%), but ultimately appear to have a relatively transient effect (6.6–11.7 months) (Dempke et al., 2015). Currently, radiotherapy appears to have better results than EGFR-TKI inhibitors and combining these treatments does not have an additive effect (Chamberlain et al., 2017; Dempke et al., 2015). However, more recent results out of the FLAURA trial show sustained effects with osimertinib (PFS 18.9 months), which is a third-generation EGFR-TKI inhibitor. In addition to improved PFS, the FLAURA trial found an a death hazard ratio that favored treatment with this biologic (HR: 0.63, p = 0.0068) (Vansteenkiste et al., 2017). This agent has engendered considerable optimism due to its modest side-effect profile and strong efficacy, but much of the support has yet to be substantiated outside of its initial trial (Soria et al., 2018).

Work with the ALK tyrosine kinase inhibitors, small molecule drugs that inhibit the constitutive tyrosine kinase activity that results from EML4 and ALK chromosomal fusions, has demonstrated good effects in NSCLC brain metastases. Crizotinib and alectinib have each demonstrated treatment benefit, albeit their use is limited to ALK fusion-positive cancers, which are remarkably rare (3–7% of NSCLC have this fusion) (Shaw et al., 2013; Peters et al., 2017; Pillai and Ramalingam, 2012). Peters et al. determined that alectinib, in particular, appears to have good CNS effect with 17/21 patients having a measurable response and 8/21 having a complete response (Peters et al., 2017). Alectinib will likely take the lead in ALK + NSCLC metastasis therapy and, due to the CNS efficacy, this agent often allows patients to defer surgical resection of their CNS metastases (Gadgeel et al., 2016; Tran and Klempner, 2017). The results of other notable studies on these agents have been organized into a table (Table 1) (Ceresoli, 2004; Iuchi et al., 2013; Welsh et al., 2013; Wu et al., 2013; Costa et al., 2015; Solomon et al., 2016). Lastly, there has been growing interest in the use of PD1-blocking antibodies in NSCLC (Passiglia et al., 2018). This therapy first found success in metastatic melanoma, but clinical trials with Pembrolizumab have yielded intriguing results for patients suffering from intracranial NSCLC metastases as well. Goldberg et al. performed a phase 2 clinical trial () on patients with intracranial metastases due to NSCLC or melanoma (Goldberg et al., 2016). Inclusion criteria for this study included positive PD1 staining, no neurological symptoms and no treatment with glucocorticoids. Eighteen NSCLC patients met these criteria and 6 (33.3%) had durable intracranial response (greater than 1 year). None of the NSCLC patients had neurological side-effects, but colitis and pneumonitis were notable treatment-ending outcomes. Lastly, it is worth noting that as our understanding of oncogenes and cancer genetics expands, so too does the role of more refined immunomodulator selection(Passiglia et al., 2018). However, the optimal dosage, delivery and drugs is still a source of active research (Frega et al., 2018).

Table 1.

Additional Studies performed on Non-Small Cell Lung Carcinoma (NSCLC).

Mechanism Agent Paper, Study Type Intracranial Response Impression/Effect Notes Toxicity
EGFR tyrosine kinase inhibitor Gefitinib Ceresoli et al., 2004
Clinical Trial
Response Rate: 10%
Median PFS: 3 mo
Disease control rate higher in patients pre-treated with WBRT and with adenocarcinoma. Did not enroll based on EGFR Status
Disease control rate higher in patients pre-treated with WBRT and with adenocarcinoma.
Grade 1 and 2 skin toxicity (24%), Diarrhea (10%)
Iuchi et al., 2013
Clinical Trial
Response Rate: 88%
Median PFS: 14.5 mo
Brain metastases respond favorably to gefitinib regardless of radiation if EGFR mutation is positive. Only included patients with EGFR mutations
Exon 19 deletion associated with better outcomes when treated with gefitinib.
Grade ≥4 toxicity: 0
Erlotinib Welsh et al.,2013
Clinical Trial
Response Rate: 86%
Median PFS: 11.8 mo
EGFR wild type: 9.3 mo
EGFR mutation: 19.1 mo
Erlotinib + WBRT is well tolerated and has a favorable response rate. Eligible patients: NSCLC with brain metastases regardless of EGFR status
Erlotinib + WBRT
Grade 3 rash requiring dose reduction: n = 3
Grade ≥4 toxicity: 0
Wu et al., 2013
Clinical Trial
Response Rate 58.3%
Median PFS: 10.1 mo
EGFR mutation positive: 15.2 mo
EGFR wild type: 4.4
Erlotinib is active and well tolerated in this patient population. Eligible patients: asymptomatic brain metastases without extracranial progressive disease post first-line platinum doublet chemotherapy Rash: 77.1%
Paronychia: 20.8%
Hyperbilirubinemia: 16.7%
Diarrhea: 14.6%
ALK tyrosine kinase inhibitor Crizotinib Costa et al.,2015
Retrospective
Review
Response Rate: 56%
Median PFS: 7 mo
Those who have systemic disease control are more likely to have intracranial disease control at 12 weeks. Eligible patients: advanced ALK-rearranged NSCLC already enrolled in clinical trial PROFILE 1005 or 1007 and randomly assigned to crizotinib retrospectively reviewed Phase III PROFILE 1014 study in ALK-positive NSCLC N/A
Solomon et al.,2016
Clinical Trial
Response Rate: 85%
Median PFS: 9 mo
Crizotinib + chemotherapy have higher intracranial disease control rate in patients with treated brain metastases. Safety comparable to patients without CNS metastases

In summary, SCLC and NSCLC are different entities, but systemic drugs can play a role for either of them. The role of systemic therapy in SCLC is complicated by the fact that it does not appear to prolong survival. Short of this aim, treatment may still have value as palliation or to delay surgery. However, conventional chemotherapy regimens, which SCLC responds to, have significant patient burden and are unlikely to improve quality of life – whole brain radiotherapy is likely the best option. This narrative is quite different for NSCLC, which is resistant to conventional agents, but may respond to well-selected targeted therapy. The positive findings with osimertinib (effective in EGFR-mutated cancers) are very intriguing. This agent induced enduring tumor response and minimal side-effects. A similar narrative appears to be unfolding for pembrolizumab (effective in PD-1 over-expressing cancers), which had a sustained response and favorable side-effect profile. For patients with EGFR-mutated or PD-1 overexpressing tumors, these agents can be considered.

3.2. Breast Cancer

Breast cancer is the second leading cause of metastatic cancer to the CNS and a growing number of recurrences are identified in the brain, even when no cancer burden is identified elsewhere in the body (Lassman and DeAngelis, 2003; Schouten et al., 2002). Breast cancer has many possible routes of therapy, and there is not an obvious regimen that is recommended to all patients. Regardless, a number of chemotherapeutics have shown efficacy in reducing the tumor size of metastatic breast cancer in the brain (Vick et al., 1977; Lin, 2013). Despite this responsiveness, first-line treatment of CNS metastases from breast cancer is typically stereotactic radiosurgery (SRS), whole brain radiotherapy and/or surgery – to date, there are no systemic therapies FDA-approved for treatment of CNS metastases from breast cancer (Venur and Leone, 2016). As a result, the tumor-shrinking effects of chemotherapy tend to be observed as an additional benefit of chemotherapy for other indications (Vick et al., 1977; Rosner et al., 1986). However, the branch of breast oncology has identified many unique targets in breast cancer that may be of use for CNS metastases in the future.

Endocrine modulating drugs have played a role in treating breast cancer since the 1960s (Venur and Leone, 2016; Salvati et al., 1993a). For tumors that express the estrogen receptor (ER), these agents can have marked effects on tumor growth. However, these effects do not appear to be carried over to CNS metastases (Venur and Leone, 2016; Burton, 2004). It is theorized that by the time breast metastases are identified in the CNS that they may have lost their ER expression (Salvati et al., 1993a; Burton, 2004; Salvati et al., 1993b). As a result, underwhelming effects are seen when these drugs are given. Despite a few small case studies that have identified tumor size reduction, the overall consensus is that endocrine modulation is not a suitable approach to CNS disease (Venur and Leone, 2016; Burton, 2004; Bousquet et al., 2016).

The history of breast cancer is also notable for the discovery of HER-2/neu – a potent transmembrane oncogene in the epidermal growth factor receptor (EGFR) family that is overexpressed in 30% of breast cancer patients (Lin, 2013). In the 1990s clinical trials validated this gene as an actionable target and trastuzumab became an essential drug in the management of breast cancers with Her-2/neu upregulation. In 2001 Slamon et al. demonstrated a 20% risk reduction of death in patients treated with trastuzumab in addition to standard of care cytotoxic therapy (Slamon et al., 2001). While the addition of this single agent was of great benefit to patients overall, this drug appears to have relatively minor effects on CNS metastases (Slamon et al., 2001; Hudis, 2007). However, enthusiasm for this agent has resurfaced due to the development of trastuzumab-emtansine (T-DM1), which is trastuzumab linked to the cytotoxic tubule inhibitor emtansine (Escrivá-de-Romaní et al., 2018). A growing body of case series and small cohort studies have shown a reduction in brain metastases and prolonged progression-free survival (Ricciardi et al., 2018; Keith et al., 2016; Okines et al., 2017; Bartsch et al., 2015). Most intriguing is the work by Krop et al. who performed a retrospective analysis of the phase 3 EMILIA trial and found significant survival benefit for T-DM1 when compared to capecitabine-lapatinib (HR = 0.38; P = 0.008; median, 26.8 versus 12.9 months). However, as a retrospective review, these findings are not altogether convincing and follow-up studies are needed. The role that this drug will play in the management of brain metastases remains to be seen, but it has shown great results thus far. Recent work with lapatinib, which acts as a tyrosine kinase inhibitor of the EGFR family, including Her-2/neu, has yielded more promising results against CNS disease (Petrelli et al., 2017). In 2013 the LANDSCAPE trial, which used lapatinib in combination with capecitabine as first-line therapy, demonstrated encouraging potential for this drug (Lin et al., 2009; Bachelot et al., 2013). This work found that 29/44 (65.9%) of patients had an objective response in CNS tumor size (> 50% tumor size reduction) without new neurological deficits (Bachelot et al., 2013). While this study demonstrated clear CNS effects, there is still a need for a randomized clinical trial to determine if this finding translates to a real survival benefit. It also remains unclear how much therapeutic effect is attributable to lapatinib as opposed to the effect of capecitabine alone. Currently, further work is needed to tease apart the effects of each of these drugs and establish the value of adding lapatinib.

In summary, the narrative of systemic therapy for breast cancer is different than the story for lung cancer. With SCLC and NSCLC alike, agents that work on extracranial metastases appear to work on intracranial tumors as well, but breast cancer does not have this symmetry. This is likely due to mutations that breast cancer manifests in order to invade the BBB. At this time, there is no convincing systemic therapy for breast cancer brain metastases (Kodack et al., 2015). If breast cancer brain metastases do indeed have unique mutations, it is likely that specific drugs will have to discovered to combat them.

3.3. Melanoma

Melanoma is an exceptionally rare cancer, but it is the third most common cause of brain metastases (Johnson and Young, 1996). This malignancy has an atypical predilection for the CNS and up to 60% of patients diagnosed with stage 4 melanoma will experience a CNS metastasis (Johnson and Young, 1996). Much of the morbidity ascribed to melanoma is due to neurological damage caused by tumor spread, and CNS metastasis is the worst prognostic indicator in melanoma (Chukwueke et al., 2016). Like many other malignancies, melanoma that has spread to the CNS is partially treated with surgical resection or radiotherapy (especially stereotactic radiosurgery) (Chukwueke et al., 2016). However, melanoma treatment has seen a recent surge in novel therapeutics that also appear to be effective in treating CNS metastases

Two checkpoint inhibitors, nivolumab and pembrolizumab, are notable for their effectiveness in metastatic melanoma (Scott, 2015; Larkin et al., 2015). These monoclonal antibodies are directed against programmed cell death protein 1 (PD-1), a molecule upregulated in cancer that suppresses the immune response. These drugs prevent PD-1 from interacting with its corresponding T-cell ligand (PDL-1) and thus cancer cells are unable to suppress the cytotoxic T-cell response. These PD-1 inhibitors appear to act best in concert with another set of monoclonals, the cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitors (Buchbinder and Desai, 2016). CTLA-4 is expressed on T-cells and interacts with ligands on dendritic cells to prevent immune responses. In the process of establishing immune quiescence, dendritic cells present tumor antigens simultaneously with the CTLA-4 ligands (CD80 and/or CD86) and immune tolerance is produced. CTLA4 antibodies act to disrupt dendritic cells inhibiting T-cell activation in the context of tumor antigens – in short, they work by inhibiting immune tolerance to cancer markers (Buchbinder and Desai, 2016). Without these antibodies, dendritic cells inhibit T-cell activation against cancer antigens and the immune system does not respond to the malignancy (Buchbinder and Desai, 2016). The combined PD-1 and CTLA-4 blockade has had considerable success, both in the CNS and outside of it. The Checkmate 204 study, which looked at asymptomatic brain metastases, showed a 55% intracranial overall response rate (ORR) and a similar extracranial ORR of 49% (Tawbi et al., 2017). Sixty-five percent of patients had PFS at 6 months (Tawbi et al., 2017). The Australian ABC trial, which was similar to Checkmate 204 in structure, reported similarly outstanding results. This team had an intracranial ORR of 42%, an extracranial ORR of 48%, and a 6 month PFS of 46% (Long et al., 2017). This project primarily looked at patients with asymptomatic CNS metastases and there is not yet a comprehensive study identifying the benefit of these drugs in symptomatic patients. The median survival has not been identified yet, which signifies how recent these studies are and how much promise still remains in this approach.

Additional molecular targets for melanoma are enzymes in the mitogen-activated protein kinase (MAPK) pathway (Chappell et al., 2011). Drugs specifically targeting BRAF and MEK have shown independent efficacy, and combining therapy to target both of these proteins simultaneously has shown remarkable benefit (Chappell et al., 2011). The major limitation of this approach is that the more robust findings are present only in patients with BRAF mutations, namely the activating BRAFV600E or BRAFV600 K mutations (Marranci et al., 2017; Shtivelman et al., 2014). Ultimately, about 50% of patients will have a BRAF mutation, most of which are BRAFV600E. The BREAK-MB trial was the first to assess the impact that a BRAF-inhibitor, dabrafenib, had in brain metastases. This study found that 44 (58%) of asymptomatic patients with BRAF600E mutations had an intracranial response(Long et al., 2012). Impressively, even patients that were less ideal candidates due to other BRAF mutations, prior therapy, and/or neurological symptoms also had response rates ranging from 44 to 59% (Long et al., 2012).Additionally, this work found that, as a single agent, dabrafenib had a relatively modest duration of effect and it was posited that a more enduring effect could be achieved with multiple agents. The COMBI-MB trial, a multi-institutional study that combined dabrafenib and trametinib (a MEK inhibitor), explored the effects of these drugs in brain metastases(Long et al., 2012; Falchook et al., 2012; Davies et al., 2017). The median response time ranged from 4.5 to 8.3 months, depending on the patients’ clinical profiles at the onset of the trial(Long et al., 2012). The treatment effect was always expected to be limited, but it was notable that intracranial response was less than the reported extracranial benefit(Long et al., 2012; Falchook et al., 2012; Davies et al., 2017). This raises the possibility that melanoma that metastasizes into the CNS evolves altered molecular targets compared to the primary tumor, or that these agents are exceptionally poor at crossing even a diseased BBB(Long et al., 2012). Regardless of the explanation, MAPK targeted dual therapy is able to induce broad, if ephemeral, remission in CNS metastases and has a favorable safety profile(Long et al., 2012, 2012; Falchook et al., 2012; Davies et al., 2017).

4. Conclusion

A key hallmark of cancer is its ability to spread and inhabit distant tissue (Hanahan and Weinberg, 2011). Malignancy that spreads to the brain can be exceptionally problematic and cause a myriad of neurological sequelae. Tumors that have migrated to the CNS may be treated with surgical resection or radiotherapy, but some may respond to systemically administered chemotherapy as well. Cytotoxic agents have historically induced modest reduction in tumor mass and will likely have a role in the management of CNS metastases alongside surgery and radiation. Many novel agents have entered the realm of CNS metastasis therapy and have expanded the options for treating these devastating tumors. Despite these advancements, there is still a great need for treatments that induce robust and sustained effects on CNS metastases.

Acknowledgments

Funding

Jonathan Rick and Ankush Chandra are supported by the Howard Hughes Medical Institute (HHMI).

Footnotes

Ethics statement

No experiments were performed on human or animal subjects in the generation of this manuscript.

Declaration of Competing Interest

The authors declare no conflict of interest.

References

  1. Davey P, 2002. Brain metastases: treatment options to improve outcomes. CNS Drugs 16, 325–338. (accessed January 1, 2018). http://www.ncbi.nlm.nih.gov/pubmed/11994022. [DOI] [PubMed] [Google Scholar]
  2. Nieder C, Mehta MP, Geinitz H, Grosu AL, 2018. Prognostic and predictive factors in patients with brain metastases from solid tumors: a review of published nomograms. Crit. Rev. Oncol. Hematol 126, 13–18. 10.1016/j.critrevonc.2018.03.018. [DOI] [PubMed] [Google Scholar]
  3. Lassman AB, DeAngelis LM, 2003. Brain metastases. Neurol. Clin 21, 1–23. (accessed January 7, 2018) vii. http://www.ncbi.nlm.nih.gov/pubmed/12690643. [DOI] [PubMed] [Google Scholar]
  4. Nayak L, Lee EQ, Wen PY, 2012. Epidemiology of brain metastases. Curr. Oncol. Rep 14, 48–54. 10.1007/s11912-011-0203-y. [DOI] [PubMed] [Google Scholar]
  5. Sandru A, Voinea S, Panaitescu E, Blidaru A, 2014. Survival rates of patients with metastatic malignant melanoma. J. Med. Life 7, 572–576. (accessed January 20, 2018). http://www.ncbi.nlm.nih.gov/pubmed/25713625. [PMC free article] [PubMed] [Google Scholar]
  6. Leone JP, Leone BA, 2015. Breast cancer brain metastases: the last frontier. Exp. Hematol. Oncol 4, 33 10.1186/s40164-015-0028-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Niemiec M, Głogowski M, Tyc-Szczepaniak D, Wierzchowski M, Kępka L, 2011. Characteristics of long-term survivors of brain metastases from lung cancer. Reports Pract. Rep. Pract. Oncol. Radiother 16, 49–53. 10.1016/J.RPOR.2011.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJB, Belanger K, Brandes AA, Marosi C, Bogdahn U, Curschmann J, Janzer RC, Ludwin SK, Gorlia T, Allgeier A, Lacombe D, Cairncross JG, Eisenhauer E, Mirimanoff RO, 2005. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N. Engl. J. Med 352, 987–996. 10.1056/NEJMoa043330. [DOI] [PubMed] [Google Scholar]
  9. Stewart DJ, 1994. A critique of the role of the blood-brain barrier in the chemotherapy of human brain tumors. J. Neurooncol 20, 121–139. 10.1007/BF01052723. [DOI] [PubMed] [Google Scholar]
  10. Zhang RD, Price JE, Fujimaki T, Bucana CD, Fidler IJ, 1992. Differential permeability of the blood-brain barrier in experimental brain metastases produced by human neoplasms implanted into nude mice. Am. J. Pathol 141, 1115–1124. (accessed January 1, 2018). http://www.ncbi.nlm.nih.gov/pubmed/1443046. [PMC free article] [PubMed] [Google Scholar]
  11. Donelli MG, Zucchetti M, D’Incalci M, 1992. Do anticancer agents reach the tumor target in the human brain? Cancer Chemother. Pharmacol 30, 251–260. (accessed January 1, 2018). http://www.ncbi.nlm.nih.gov/pubmed/1643692. [DOI] [PubMed] [Google Scholar]
  12. Lesser GJ, 1996. Chemotherapy of cerebral metastases from solid tumors. Neurosurg. Clin. N. Am 7, 527–536. (accessed January 1, 2018). http://www.ncbi.nlm.nih.gov/pubmed/8823780. [PubMed] [Google Scholar]
  13. Chamberlain MC, Baik CS, Gadi VK, Bhatia S, Chow LQM, 2017. Systemic therapy of brain metastases: non–small cell lung cancer, breast cancer, and melanoma. Neuro. Oncol 19, i1–i24. 10.1093/neuonc/now197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Whitsett TG, Inge LJ, Dhruv HD, Cheung PY, Weiss GJ, Bremner RM, Winkles JA, Tran NL, 2013. Molecular determinants of lung cancer metastasis to the central nervous system. Transl. Lung Cancer Res 2, 273–283. 10.3978/j.issn.2218-6751.2013.03.12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Barnholtz-Sloan JS, Sloan AE, Davis FG, Vigneau FD, Lai P, Sawaya RE, 2004. Incidence proportions of brain metastases in patients diagnosed (1973 to 2001) in the metropolitan detroit Cancer surveillance system. J. Clin. Oncol 22, 2865–2872. 10.1200/JCO.2004.12.149. [DOI] [PubMed] [Google Scholar]
  16. Herbst RS, Heymach JV, Lippman SM, 2008. Lung Cancer. N. Engl. J. Med 359, 1367–1380. 10.1056/NEJMra0802714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Chen G, Huynh M, Chen A, Fehrenbacher L, Gandara D, Lau D, 2008. Chemotherapy for brain metastases in small-cell lung Cancer. Clin. Lung Cancer 9, 35–38. 10.3816/CLC.2008.n.006. [DOI] [PubMed] [Google Scholar]
  18. Postmus PE, Smit EF, 1999. Chemotherapy for brain metastases of lung cancer: a review. Ann. Oncol 10, 753–759. 10.1023/A:1008318515795. [DOI] [PubMed] [Google Scholar]
  19. Twelves CJ, Souhami RL, Harper PG, Ash CM, Spiro SG, Earl HM, Tobias JS, Quinn H, Geddes DM, 1990. The response of cerebral metastases in small cell lung cancer to systemic chemotherapy. Br. J. Cancer 61, 147–150. (accessed January 16, 2018). http://www.ncbi.nlm.nih.gov/pubmed/2153393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Kristjansen PE, Soelberg Sørensen P, Skov Hansen M, Hansen HH, 1993. 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. Off. J. Eur. Soc. Med. Oncol 4, 579–583. (accessed January 16, 2018). http://www.ncbi.nlm.nih.gov/pubmed/8395873. [DOI] [PubMed] [Google Scholar]
  21. Reveiz L, Rueda J-R, Cardona AF, 2012. Chemotherapy for brain metastases from small cell lung cancer. Cochrane Database Syst. Rev, CD007464 10.1002/14651858.CD007464.pub2. [DOI] [PMC free article] [PubMed]
  22. Dempke WCM, Edvardsen K, Lu S, Reinmuth N, Reck M, Inoue A, 2015. Brain metastases in NSCLC - are TKIs changing the treatment strategy? Anticancer Res 35, 5797–5806. (accessed January 17, 2018). http://www.ncbi.nlm.nih.gov/pubmed/26504000. [PubMed] [Google Scholar]
  23. Vansteenkiste J, Reungwetwattana T, Nakagawa K, Cho BC, Dols MAC, Cho EK, Bertolini A, Bohnet S, Zhou C, Lee KH, Nogami N, Okamoto I, Leighl N, Hodge R, McKeown A, Brown AP, Rukazenkov Y, Ramalingam S, 2017. LBA5CNS response to osimertinib vs standard of care (SoC) EGFR-TKI as first-line therapy in patients (pts) with EGFR-TKI sensitising mutation (EGFRm)-positive advanced non-small cell lung cancer (NSCLC): data from the FLAURA study. Ann. Oncol 28 10.1093/annonc/mdx729.007. [DOI] [Google Scholar]
  24. Soria J-C, Ohe Y, Vansteenkiste J, Reungwetwattana T, Chewaskulyong B, Lee KH, Dechaphunkul A, Imamura F, Nogami N, Kurata T, Okamoto I, Zhou C, Cho BC, Cheng Y, Cho EK, Voon PJ, Planchard D, Su W-C, Gray JE, Lee S-M, Hodge R, Marotti M, Rukazenkov Y, Ramalingam SS, 2018. Osimertinib in untreated EGFR -Mutated advanced non–Small-Cell lung Cancer. N. Engl. J. Med 378, 113–125. 10.1056/NEJMoa1713137. [DOI] [PubMed] [Google Scholar]
  25. Shaw AT, Kim D-W, Nakagawa K, Seto T, Crinó L, Ahn M-J, De Pas T, Besse B, Solomon BJ, Blackhall F, Wu Y-L, Thomas M, O’Byrne KJ, Moro-Sibilot D, Camidge DR, Mok T, Hirsh V, Riely GJ, Iyer S, Tassell V, Polli A, Wilner KD, Jänne PA, 2013. Crizotinib versus chemotherapy in advanced ALK -Positive lung Cancer. N. Engl. J. Med 368, 2385–2394. 10.1056/NEJMoa1214886. [DOI] [PubMed] [Google Scholar]
  26. Peters S, Camidge DR, Shaw AT, Gadgeel S, Ahn JS, Kim D-W, Ou S-HI, Pérol M, Dziadziuszko R, Rosell R, Zeaiter A, Mitry E, Golding S, Balas B, Noe J, Morcos PN, Mok T, 2017. ALEX trial investigators, alectinib versus crizotinib in untreated ALK -Positive non–Small-Cell lung Cancer. N. Engl. J. Med 377, 829–838. 10.1056/NEJMoa1704795. [DOI] [PubMed] [Google Scholar]
  27. Pillai RN, Ramalingam SS, 2012. The biology and clinical features of non–small cell lung cancers with EML4-ALK translocation. Curr. Oncol. Rep 14, 105–110. 10.1007/s11912-012-0213-4. [DOI] [PubMed] [Google Scholar]
  28. Gadgeel SM, Shaw AT, Govindan R, Gandhi L, Socinski MA, Camidge DR, De Petris L, Kim D-W, Chiappori A, Moro-Sibilot DL, Duruisseaux M, Crino L, De Pas T, Dansin E, Tessmer A, Yang JC-H, Han J-Y, Bordogna W, Golding S, Zeaiter A, Ou S-HI, 2016. Pooled analysis of CNS response to alectinib in two studies of pretreated patients with ALK-Positive non-small-Cell lung Cancer. J. Clin. Oncol 34, 4079–4085. 10.1200/JCO.2016.68.4639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Tran PN, Klempner SJ, 2017. ALK on my mind: alectinib takes an early lead in managing intracranial disease in non-small cell lung cancer with ALK rearrangements. Ann. Transl. Med 5 10.21037/atm.2017.03.47 173–173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Ceresoli GL, 2004. Gefitinib in patients with brain metastases from non-small-cell lung cancer: a prospective trial. Ann. Oncol 15, 1042–1047. 10.1093/annonc/mdh276. [DOI] [PubMed] [Google Scholar]
  31. Iuchi T, Shingyoji M, Sakaida T, Hatano K, Nagano O, Itakura M, Kageyama H, Yokoi S, Hasegawa Y, Kawasaki K, Iizasa T, 2013. Phase II trial of gefitinib alone without radiation therapy for Japanese patients with brain metastases from EGFR-mutant lung adenocarcinoma. Lung Cancer 82, 282–287. 10.1016/j.lungcan.2013.08.016. [DOI] [PubMed] [Google Scholar]
  32. Welsh JW, Komaki R, Amini A, Munsell MF, Unger W, Allen PK, Chang JY, Wefel JS, McGovern SL, Garland LL, Chen SS, Holt J, Liao Z, Brown P, Sulman E, Heymach JV, Kim ES, Stea B, 2013. 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 31, 895–902. 10.1200/JCO.2011.40.1174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Wu Y-L, Zhou C, Cheng Y, Lu S, Chen G-Y, Huang C, Huang Y-S, Yan H-H, Ren S, Liu Y, Yang J-J, 2013. Erlotinib as second-line treatment in patients with advanced non-small-cell lung cancer and asymptomatic brain metastases: a phase II study (CTONG-0803). Ann. Oncol 24, 993–999. 10.1093/annonc/mds529. [DOI] [PubMed] [Google Scholar]
  34. Costa DB, Shaw AT, Ou S-HI, Solomon BJ, Riely GJ, Ahn M-J, Zhou C, Shreeve SM, Selaru P, Polli A, Schnell P, Wilner KD, Wiltshire R, Camidge DR, Crinò L, 2015. Clinical experience with crizotinib in patients with advanced ALK -Rearranged non–Small-Cell lung Cancer and brain metastases. J. Clin. Oncol 33, 1881–1888. 10.1200/JCO.2014.59.0539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Solomon BJ, Cappuzzo F, Felip E, Blackhall FH, Costa DB, Kim DW, Nakagawa K, Wu YL, Mekhail T, Paolini J, Tursi J, Usari T, Wilner KD, Selaru P, Mok TSK, 2016. Intracranial efficacy of crizotinib versus chemotherapy in patients with advanced ALK-positive non-small-cell lung cancer: results from PROFILE 1014. J. Clin. Oncol 34, 2858–2865. 10.1200/JCO.2015.63.5888. [DOI] [PubMed] [Google Scholar]
  36. Passiglia F, Caglevic C, Giovannetti E, Pinto J, Manca P, Taverna S, Listì A, Gil-Bazo I, Raez L, Russo A, Rolfo C, 2018. Primary and metastatic brain cancer genomics and emerging biomarkers for immunomodulatory cancer treatment. Semin. Cancer Biol 52, 259–268. 10.1016/j.semcancer.2018.01.015. [DOI] [PubMed] [Google Scholar]
  37. Goldberg SB, Gettinger SN, Mahajan A, Chiang AC, Herbst RS, Sznol M, Tsiouris AJ, Cohen J, Vortmeyer A, Jilaveanu L, Yu J, Hegde U, Speaker S, Madura M, Ralabate A, Rivera A, Rowen E, Gerrish H, Yao X, Chiang V, Kluger HM, 2016. Pembrolizumab for patients with melanoma or non-small-cell lung cancer and untreated brain metastases: early analysis of a non-randomised, open-label, phase 2 trial. Lancet Oncol 17, 976–983. 10.1016/S1470-2045(16)30053-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Frega S, Bonanno L, Guarneri V, Conte P, Pasello G, 2018. Therapeutic perspectives for brain metastases in non-oncogene addicted non-small cell lung cancer (NSCLC): Towards a less dismal future? Crit. Rev. Oncol. Hematol 128, 19–29. 10.1016/j.critrevonc.2018.05.013. [DOI] [PubMed] [Google Scholar]
  39. Schouten LJ, Rutten J, Huveneers HAM, Twijnstra A, 2002. Incidence of brain metastases in a cohort of patients with carcinoma of the breast, colon, kidney, and lung and melanoma. Cancer 94, 2698–2705. (accessed January 7, 2018). http://www.ncbi.nlm.nih.gov/pubmed/12173339. [DOI] [PubMed] [Google Scholar]
  40. Vick NA, Khandekar JD, Bigner DD, 1977. Chemotherapy of brain tumors. Arch. Neurol 34, 523–526. (accessed January 17, 2018). http://www.ncbi.nlm.nih.gov/pubmed/889492. [DOI] [PubMed] [Google Scholar]
  41. Lin NU, 2013. Breast cancer brain metastases: new directions in systemic therapy. Ecancermedicalscience 7, 307 10.3332/ecancer.2013.307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Venur VA, Leone JP, 2016. Targeted therapies for brain metastases from breast Cancer. Int. J. Mol. Sci 17 10.3390/ijms17091543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Rosner D, Nemoto T, Lane WW, 1986. Chemotherapy induces regression of brain metastases in breast carcinoma. Cancer 58, 832–839. (accessed January 17, 2018). http://www.ncbi.nlm.nih.gov/pubmed/3755076. [DOI] [PubMed] [Google Scholar]
  44. Salvati M, Cervoni L, Innocenzi G, Bardella L, 1993a. Prolonged stabilization of multiple and single brain metastases from breast cancer with tamoxifen. Report of three cases. Tumori 79, 359–362. (accessed January 17, 2018). http://www.ncbi.nlm.nih.gov/pubmed/8116083. [DOI] [PubMed] [Google Scholar]
  45. Burton A, 2004. High incidence of brain metastases with trastuzumab treatment. Lancet Oncol 5, 523 10.1016/S1470-2045(04)01558-X. [DOI] [PubMed] [Google Scholar]
  46. Salvati M, Cervoni L, Innocenzi G, Bardella L, 1993b. Prolonged stabilization of multiple and single brain metastases from breast cancer with tamoxifen. Report of three cases. Tumori 79, 359–362. (accessed January 18, 2018). http://www.ncbi.nlm.nih.gov/pubmed/8116083. [DOI] [PubMed] [Google Scholar]
  47. Bousquet G, Darrouzain F, de Bazelaire C, Ternant D, Barranger E, Winterman S, Madelaine-Chambin I, Thiebaut J-B, Polivka M, Paintaud G, Culine S, Janin A, 2016. Intrathecal trastuzumab halts progression of CNS metastases in breast Cancer. J. Clin. Oncol 34, e151–5. 10.1200/JCO.2012.44.8894. [DOI] [PubMed] [Google Scholar]
  48. Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, Fleming T, Eiermann W, Wolter J, Pegram M, Baselga J, Norton L, 2001. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast Cancer That overexpresses HER2. N. Engl. J. Med 344, 783–792. 10.1056/NEJM200103153441101. [DOI] [PubMed] [Google Scholar]
  49. Hudis CA, 2007. Trastuzumab — mechanism of action and use in clinical practice. N. Engl. J. Med 357, 39–51. 10.1056/NEJMra043186. [DOI] [PubMed] [Google Scholar]
  50. Escrivá-de-Romaní S, Arumí M, Bellet M, Saura C, 2018. HER2-positive breast cancer: Current and new therapeutic strategies. Breast 39, 80–88. 10.1016/j.breast.2018.03.006. [DOI] [PubMed] [Google Scholar]
  51. Ricciardi GRR, Russo A, Franchina T, Schifano S, Mastroeni G, Santacaterina A, Adamo V, 2018. Efficacy of T-DM1 for leptomeningeal and brain metastases in a HER2 positive metastatic breast cancer patient: new directions for systemic therapy - a case report and literature review. BMC Cancer 18, 97 10.1186/s12885-018-3994-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Keith KC, Lee Y, Ewend MG, Zagar TM, Anders CK, 2016. Activity of TRASTUZUMAB-EMTANSINE (TDM1) in HER2-POSITIVE breast cancer brain metastases: a case series. Cancer Treat. Commun 7, 43–46. 10.1016/j.ctrc.2016.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Okines A, Irfan T, Khabra K, Smith I, O’Brien M, Parton M, Noble J, Stanway S, Somaiah N, Ring A, Johnston S, Turner N, 2017. Development and responses of brain metastases during treatment with trastuzumab emtansine (T-DM1) for HER2 positive advanced breast cancer: a single institution experience. Breast J 10.1111/tbj.12906. [DOI] [PubMed]
  54. Bartsch R, Berghoff AS, Vogl U, Rudas M, Bergen E, Dubsky P, Dieckmann K, Pinker K, Bago-Horvath Z, Galid A, Oehler L, Zielinski CC, Gnant M, Steger GG, Preusser M, 2015. Activity of T-DM1 in Her2-positive breast cancer brain metastases. Clin. Exp. Metastasis 32, 729–737. 10.1007/s10585-015-9740-3. [DOI] [PubMed] [Google Scholar]
  55. Petrelli F, Ghidini M, Lonati V, Tomasello G, Borgonovo K, Ghilardi M, Cabiddu M, Barni S, 2017. The efficacy of lapatinib and capecitabine in HER-2 positive breast cancer with brain metastases: a systematic review and pooled analysis. Eur. J. Cancer 84, 141–148. 10.1016/j.ejca.2017.07.024. [DOI] [PubMed] [Google Scholar]
  56. Lin NU, Diéras V, Paul D, Lossignol D, Christodoulou C, Stemmler H-J, Roché H, Liu MC, Greil R, Ciruelos E, Loibl S, Gori S, Wardley A, Yardley D, Brufsky A, Blum JL, Rubin SD, Dharan B, Steplewski K, Zembryki D, Oliva C, Roychowdhury D, Paoletti P, Winer EP, 2009. Multicenter phase II study of lapatinib in patients with brain metastases from HER2-positive breast cancer. Clin. Cancer Res 15, 1452–1459. 10.1158/1078-0432.CCR-08-1080. [DOI] [PubMed] [Google Scholar]
  57. Bachelot T, Romieu G, Campone M, Diéras V, Cropet C, Dalenc F, Jimenez M, Le Rhun E, Pierga J-Y, Gonçalves A, Leheurteur M, Domont J, Gutierrez M, Curé H, Ferrero J-M, Labbe-Devilliers C, 2013. 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 14, 64–71. 10.1016/S1470-2045(12)70432-1. [DOI] [PubMed] [Google Scholar]
  58. Kodack DP, Askoxylakis V, Ferraro GB, Fukumura D, Jain RK, 2015. Emerging strategies for treating brain metastases from breast Cancer. Cancer Cell 27, 163–175. 10.1016/j.ccell.2015.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Johnson JD, Young B, 1996. Demographics of brain metastasis. Neurosurg. Clin. N. Am 7, 337–344. (accessed January 18, 2018). http://www.ncbi.nlm.nih.gov/pubmed/8823767. [PubMed] [Google Scholar]
  60. Chukwueke U, Batchelor T, Brastianos P, 2016. Management of brain metastases in patients with melanoma. J. Oncol. Pract 12, 536–542. 10.1200/JOP.2016.011882. [DOI] [PubMed] [Google Scholar]
  61. Scott LJ, 2015. Nivolumab: a review in advanced melanoma. Drugs 75, 1413–1424. 10.1007/s40265-015-0442-6. [DOI] [PubMed] [Google Scholar]
  62. Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, Schadendorf D, Dummer R, Smylie M, Rutkowski P, Ferrucci PF, Hill A, Wagstaff J, Carlino MS, Haanen JB, Maio M, Marquez-Rodas I, McArthur GA, Ascierto PA, Long GV, Callahan MK, Postow MA, Grossmann K, Sznol M, Dreno B, Bastholt L, Yang A, Rollin LM, Horak C, Hodi FS, Wolchok JD, 2015. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N. Engl. J. Med 373, 23–34. 10.1056/NEJMoa1504030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Buchbinder EI, Desai A, 2016. CTLA-4 and PD-1 pathways. Am. J. Clin. Oncol 39, 98–106. 10.1097/COC.0000000000000239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Tawbi HA-H, Forsyth PAJ, Algazi AP, Hamid O, Hodi FS, Moschos SJ, Khushalani NI, Gonzalez R, Lao CD, Postow MA, Atkins MB, Ernstoff MS, Puzanov I, Kudchadkar RR, Thomas RP, Tarhini AA, Jiang J, Avila A, Demelo S, Margolin KA, 2017. Efficacy and safety of nivolumab (NIVO) plus ipilimumab (IPI) in patients with melanoma (MEL) metastatic to the brain: results of the phase II study CheckMate 204. J. Clin. Oncol 35, 9507 10.1200/JCO.2017.35.15_suppl.9507. [DOI] [Google Scholar]
  65. Long GV, Atkinson V, Menzies AM, Lo S, Guminski AD, Brown MP, Gonzalez MM, Diamante K, Sandhu SK, Scolyer RA, Emmett L, McArthur GA, 2017. A randomized phase II study of nivolumab or nivolumab combined with ipilimumab in patients (pts) with melanoma brain metastases (mets): the Anti-PD1 Brain Collaboration (ABC). J. Clin. Oncol 35, 9508 10.1200/JCO.2017.35.15_suppl.9508. [DOI] [Google Scholar]
  66. Chappell WH, Steelman LS, Long JM, Kempf RC, Abrams SL, Franklin RA, Bäsecke J, Stivala F, Donia M, Fagone P, Malaponte G, Mazzarino MC, Nicoletti F, Libra M, Maksimovic-Ivanic D, Mijatovic S, Montalto G, Cervello M, Laidler P, Milella M, Tafuri A, Bonati A, Evangelisti C, Cocco L, Martelli AM, McCubrey JA, 2011. Ras/Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR inhibitors: rationale and importance to inhibiting these pathways in human health. Oncotarget 2, 135–164. 10.18632/oncotarget.240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Marranci A, Jiang Z, Vitiello M, Guzzolino E, Comelli L, Sarti S, Lubrano S, Franchin C, Echevarría-Vargas I, Tuccoli A, Mercatanti A, Evangelista M, Sportoletti P, Cozza G, Luzi E, Capobianco E, Villanueva J, Arrigoni G, Signore G, Rocchiccioli S, Pitto L, Tsinoremas N, Poliseno L, 2017. The land-scape of BRAF transcript and protein variants in human cancer. Mol. Cancer 16, 85 10.1186/s12943-017-0645-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Shtivelman E, Davies MQA, Hwu P, Yang J, Lotem M, Oren M, Flaherty KT, Fisher DE, 2014. Pathways and therapeutic targets in melanoma. Oncotarget 5, 1701–1752. 10.18632/oncotarget.1892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Long GV, Trefzer U, Davies MA, Kefford RF, Ascierto PA, Chapman PB, Puzanov I, Hauschild A, Robert C, Algazi A, Mortier L, Tawbi H, Wilhelm T, Zimmer L, Switzky J, Swann S, Martin A-M, Guckert M, Goodman V, Streit M, Kirkwood JM, Schadendorf D, 2012. 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 13, 1087–1095. 10.1016/S1470-2045(12)70431-X. [DOI] [PubMed] [Google Scholar]
  70. Falchook GS, Long GV, Kurzrock R, Kim KB, Arkenau TH, Brown MP, Hamid O, Infante JR, Millward M, Pavlick AC, O’Day SJ, Blackman SC, Curtis CM, Lebowitz P, Ma B, Ouellet D, Kefford RF, 2012. Dabrafenib in patients with melanoma, untreated brain metastases, and other solid tumours: a phase 1 dose-escalation trial. Lancet (London, England) 379, 1893–1901. 10.1016/S0140-6736(12)60398-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Davies MA, Saiag P, Robert C, Grob J-J, Flaherty KT, Arance A, Chiarion-Sileni V, Thomas L, Lesimple T, Mortier L, Moschos SJ, Hogg D, Márquez-Rodas I, Del Vecchio M, Lebbé C, Meyer N, Zhang Y, Huang Y, Mookerjee B, Long GV, 2017. Dabrafenib plus trametinib in patients with BRAFV600-mutant melanoma brain metastases (COMBI-MB): a multicentre, multicohort, open-label, phase 2 trial. Lancet Oncol 18, 863–873. 10.1016/S1470-2045(17)30429-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Hanahan D, Weinberg RA, 2011. Hallmarks of Cancer: the next generation. Cell 144, 646–674. 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]

RESOURCES