Skip to main content
Erschienen in: memo - Magazine of European Medical Oncology 2/2021

Open Access 22.05.2021 | short review

Brain metastases: new systemic treatment approaches

verfasst von: Ariane Steindl, MD PhD, Anna Sophie Berghoff

Erschienen in: memo - Magazine of European Medical Oncology | Ausgabe 2/2021

Summary

Background

Brain metastases (BM) still present a clinical challenge in oncology. Treatment of BM was mainly based on local approaches including neurosurgery and radiation. However, the fraction of patients with asymptomatic BM has risen over the last decade. Recent clinical trials on immune- and targeted therapies showed promising intracranial responses—especially in neurological asymptomatic status. Therefore, systemic treatment presents an emerging therapy approach specifically in patients with asymptomatic BM.

Methods

The present review highlights the recent advances in systemic therapeutic and preventive approaches in BM focusing on the main BM causing tumors: non-small cell lung cancer (NSCLC), melanoma and breast cancer.

Results

Remarkable intracranial efficacies were presented for several next-generation tyrosine kinase inhibitors (TKIs) and immune checkpoint inhibitors, especially in asymptomatic BM patients. In NSCLC, osimertinib and afatinib presented intracranial response rates over 80%. Osimertinib showed even a potential for primary BM prevention. Considerable intracranial response rates were observed for the combination of dabrafenib and trametinib in BRAF mutated melanoma BM. Combined ipilimumab and nivolumab treatment in asymptomatic melanoma BM even presented with similar intra- and extracranial response rates. In breast cancer, HER2-targeted TKIs like lapatinib in combination with chemotherapy, or trastuzumab deruxtecan monotherapy presented also notable intracranial response rates.

Conclusion

New developments in targeted and immune-modulating therapies have postulated high intracranial efficacies in patients with BM from different solid tumors. However, more BM-specific studies and BM-specific endpoints in registration trials are warranted to underscore the role of systemic treatment in patients with BM.
Hinweise
The original online version of this article was revised: The licence information was missing from this article and should have been “Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​”.
A correction to this article is available online at https://​doi.​org/​10.​1007/​s12254-022-00827-4.

Publisher’s Note

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

Background

Brain metastases (BM) are associated with a severe impact on quality of life and survival prognosis. Beside the common primary tumors causing BM including lung cancer, breast cancer and melanoma, the incidence of BM due to colon cancer, renal cell carcinoma and rare tumor types were reported to be rising resulting in BM being the most common tumor of the central nervous system [1]. However, the clinical presentation and the clinical course of BM have changed over the last decades, leading to a new field of therapeutic and preventive approaches and an improvement of prognosis in brain metastatic patients.

Neurological symptom burden in BM: inclusion in treatment decisions?

Due to wider application and availability of BM screening, the clinical presentation of BM has changed during the last decades. Analyses from a large real-life cohort uncovered a rising incidence of BM diagnosed during screening and staging procedures in absence of neurological symptoms over the last decades [1, 2]. While in the 1990s, the incidence of asymptomatic patients at BM diagnosis was 15.3%, in the period from 2000–2009 the incidence had already doubled to 31.4%. Furthermore, this subgroup of BM patients was associated with a significantly longer survival prognosis (11 vs. 7 months) and presented more frequently an extracranial progression (35.7% vs. 50.3%) as major cause of death, compared to symptomatic patients. Therefore, combined intra- and extracranial disease control as well as prevention of long-term neurocognitive toxicity has to be considered in treatment planning of asymptomatic BM patients.
Thus, the current European Association of Neuro-oncology (EANO) guidelines for BM treatment recommends the more invasive therapeutic approaches like neurosurgical resections, stereotactic radiosurgery (SRS) and whole-brain radiotherapy (WBRT) in patients with high intracranial symptom burden in need for immediate symptom relief, while initial systemic treatment should be evaluated in patients with asymptomatic BM—if intracranially active systemic treatment is available. In the event of low performance status and/or progressive systemic disease with limited survival prognosis, the use of WBRT or best supportive care should be evaluated (Fig. 1; [3, 4]). Importantly, best supportive care as the primary treatment approach should not be underrated in patients with limited survival prognosis as the QUARTZ (Quality of Life after Radiotherapy for Brain Metastases) trial—a randomized phase III trial comparing BSC with WBRT versus BSC alone for patients with BM from NSCLC—showed no significant differences in overall survival or quality of life between the groups [5].

New era of BM treatment: systemic therapies as initial treatment approach

Indeed, a major step of systemic treatment in advanced cancer was the successful introduction of immune-related therapy as well as tyrosine kinase inhibitors (TKI) in several BM-causing tumor entities during the last decades. BM-specific endpoints as well as inclusion of brain metastatic patients have been implemented in several modern trials on targeted and immune-modulating therapies, resulting in various studies presenting remarkable intracranial efficacies—especially, in patients with neurological asymptomatic disease ([4]; Table 1). In line with this, several studies also revealed an intracranial expression of predictive biomarkers—even though with heterogeneity between the sites—further supporting the evaluation of these emerging treatment approaches in metastatic brain cancer [6].
Table 1
Selected studies on the efficacies of systemic therapies in BM patients [4]
Entity
Target
Characteristics
Drug
IC RR (%)
EC RR (%)
OS (months)
Ref.
NSCLC
EGFR mutation
Asymp
Afatinib
81.8
82.1
22.4
[7, 8]
NSCLC
EGFR mutation
Asymp
Osimertinib
66–91.0
77
38.6
[5]
NSCLC
ALK translocation
Asymp
Crizotinib
40.0–71.4
53.0–55.0
Immature
[9, 10]
NSCLC
ALK translocation
Asymp
Alectinib
78.6–85.7
NG
Immature
[9, 10]
NSCLC
ALK translocation
Symp
Alectinib
54.2
NG
Immature
[9, 10]
NSCLC
ALK translocation
Asymp. + untreat./pretreat
Lorlatinib
63–66.7
NG
NG
[12, 13]
NSCLC
ALK translocation
NG
Brigatinib
78.0
NG
NR
[14]
NSCLC
PD‑1
Asymp
Pembrolizumab
33.0
33.0
7.7
[25]
NSCLC
PD‑1
Asymp
Nivolumab
9.0
11.0
7.5
[28]
NSCLC
PDL‑1
Asymp
Atezolizumab
NG
NG
13.2–16.0
 
Melanoma
BRAF mut.+MEK
Asymp./
Untreat + Pretreat
Dabrafenib + trametinib
44–58
44–75.0
10.1–24.3
[15, 16]
Melanoma
BRAF mut.+MEK
Symp./
Untreat + Pretreat
Dabrafenib + trametinib
59.0
41.0
11.5
[15, 16]
Melanoma
CTLA‑4
Asymp
Ipilimumab
16.0
14.0
7.0
[22]
Melanoma
CTLA‑4
Symp
Ipilimumab
5.0
5.0
3.7
[22]
Melanoma
PD‑1
Asymp
Pembrolizumab
22.0
22.0
NR
[23]
Melanoma
PD‑1+CTLA‑4
Asymp
Ipilimumab + Nivolumab
55.0
50.0
NR
[24, 25]
Breast Cancer
HER‑2
Asymp
Trastuzumab deruxtecan
58.0
NG
NR
[20]
Breast Cancer
HER‑2
Asymp
Lapatinib + Capecitabine
31–57
NG
17.0
[17]
Breast Cancer
HER‑2
Asymp
Trastuzumab + Pertuzumab +Docetaxel
NG
NG
56.5
[16]
Breast Cancer
HER‑2
Sympt. + Pretreat/untreat
Neratinib + Capecitabine
33–49
14–43
13.3–21.0
[18]
Breast Cancer
HER‑2
Asymp
Tucatinib + Trastuzumab +Capecitabine
40.6
NG
21.9
[19]
ALK Anaplastic lymphoma kinase, Asymp Asymptomatic brain metastasis, BRAF v-Raf murine sarcoma viral oncogene homolog B, CTLA‑4 cytotoxic T‑lymphocyte-associated protein 4, EC RR Extracranial response rate, EGR‑R Epidermal growth factor receptor, HER‑2 Human epidermal growth factor receptor 2, IC RR Intracranial response rate, NG Not given, NR Not reached, NSCLC Non-small cell lung cancer, OS Overall survival, PD‑1 Programmed cell death protein 1, Ref. References, Symp. Symptomatic brain metastasis, Treat Treated, Untreat Untreated

Tyrosine kinase inhibitors in BM treatment

Third-generation epidermal growth factor receptor (EGFR) TKIs like osimertinib presented remarkable intracranial response rates over 80% in patients with EGFR-mutated metastatic lung cancer and asymptomatic BM [79]. In contrast to first- and second-generation EGFR TKIs, osimertinib is even active in the presence of an EGFR T790 mutation—a point mutation of the EGFR, which is responsible for more than 50% of treatment resistances to first- and second-generation EGFR TKIs. Furthermore, also in ALK- and ROS1 rearranged NSCLC, only the third generation ALK inhibitors alectinib, lorlatinib and brigatinib were associated with meaningful intracranial response rates over 65% [1014]. In the registration trials of alectinib and lorlatinib even small subgroups of pretreated and symptomatic BM patients were included; however, compared to asymptomatic patients, reduced intracranial efficacies were observed [10, 12].
Similar high intracranial response rates (78.9%) were observed for combined BRAF-inhibitor dabrafenib and mitogen-activated protein kinase (MEK) inhibitor trametinib in BRAF V600E-mutated and asymptomatic melanoma BM [15]. However, in case of symptomatic BM with or without previous treatment, the intracranial response rates of combined treatment approaches were reduced (59%) [15].
The HER2 subtype in breast cancer presents the most favorable prognosis after diagnosis of BM as survival prognosis of up to 24 months and longer can be observed [1]. While in luminal and triple negative breast cancer only limited to no proven targeted therapy approaches with favorable intracranial efficacies are available, HER2-directed treatment has shown successful intracranial responses in HER2 overexpressed breast cancer BM. On the one hand, combination treatment approaches as HER2-targeted TKI like lapatinib, neratinib or tucatinib in combination with chemotherapy, and HER2-targeted antibodies like trastuzumab and pertuzumab in combination with chemotherapy have shown intracranial efficacies (intracranial response rates: 31–57%) [1619]. On the other hand, HER2-targeted antibody–drug conjugate monotherapy with the more potent trastuzumab deruxtecan, an antibody–drug conjugate composed of an anti-HER2 antibody and a cytotoxic topoisomerase I inhibitor, presented with almost identical extracranial (58%) and intracranial (62%) response rates in heavily pretreated HER2-positive metastatic breast cancer patients [20].

Immune checkpoint inhibitors in BM treatment

The PD‑1 directed immune checkpoint inhibitor pembrolizumab as monotherapy in melanoma BM presented an intracranial response rate of 22%—only about half of that seen in extracranial disease (40%) [21]. In contrast, the combined therapy of the PD‑1 inhibitor nivolumab with the CTLA‑4 inhibitor ipilimumab in asymptomatic melanoma BM showed similar intra- and extracranial response rates up to 47–55% [22]. This study also allowed the inclusion of a subgroup of symptomatic melanoma BM, which showed lower intracranial efficacy in symptomatic (clinical benefit rate: 22.2%) compared to asymptomatic (clinical benefit rate of 58.4%) BM patients [23]. However, 87% of the asymptomatic BM patients still had an ongoing response at the time of data cutoff, underscoring the more durable responses of immune directed therapy compared to BRAF/MEK inhibitor treatment in melanoma BM.
Immune checkpoint inhibitors, either as monotherapy or in combination with chemotherapy, targeting the PD‑1 (nivolumab/pembrolizumab) or the PD-L1 (atezolizumab) axis are well-established treatment approaches in metastatic NSCLC. In a phase II study of pembrolizumab in NSCLC an intracranial response rate of 33% and an extracranial response rate of 50% could be observed [21]. Furthermore, the intracranial efficacy of nivolumab was postulated as not inferior compared to the extracranial efficacy (intracranial: 9% vs. extracranial: 11%) in NSCLC with locally pretreated, active BM [24]. In addition, atezolizumab was even associated with a secondary BM preventive potential in patients with treated BM [25]. In a post hoc analysis of the OAK trial comparing the PD-L1 inhibitor atezolizumab to docetaxel as a second-line treatment in NSCLC, the median time to new BM lesions was lower with atezolizumab than with docetaxel (not reached vs. 9.5 months; HR = 0.42; 95% CI: 0.15–0.18) [25].
In breast cancer, up to now, only the IMpassion130 has presented an overall survival benefit for an immune checkpoint inhibitor-based therapy in patients with newly diagnosed, triple-negative locally advanced or metastatic breast cancer [26]. Even though brain metastatic patients have been excluded from this registration trial, a BM-specific prospective trial is currently investigating the intracranial efficacy of atezolizumab-based treatment combined with (nab‑) paclitaxel in triple-negative breast cancer patients irrespective of PD-L1 expression (NCT03483012).

Prevention of brain metastasis: the new landmark of systemic treatment?

For a long time, prophylactic cranial irradiation (PCI) presented the only possible approach to prevent BM. However, due to the unfavorable neurological side effects and the limited impact on prognosis, the use of PCI is restricted to specific indications [3]. As the treatment of symptomatic BM is challenging and the overall survival prognosis still poor, secondary and primary prevention of BM by systemic treatment came into the focus in recent years (Table 2; Fig. 2).
Table 2
Primary BM prevention in selected NSCLC studies
Primary BM prevention in NSCLC
Incidence of BM during systemic treatment
(%)
Osimertinib
3.0
Geftinib/Erlotinib
7.0
Alectinib
4.6
Crizotinib
31.5
Radiochemotherapy+Durvalumab
5.5
Radiochemotherapy+Placebo
11.0
BM Brain metastases, NSCLC Non-small cell lung cancer
In the FLAURA trial, comparing osimertinib with standard EGFR-TKI, 3% of patients without BM at baseline developed BM during the follow-up in the osimertinib group compared to 7% in the standard EGFR-TKI group. Furthermore, 19% of patients with BM at baseline developed new or progressive BM in the osimertinib group compared to 43% in the standard EGFR-TKI group [27].
Primary BM prevention of third generation ALK inhibitor alectinib compared to crizotinib was investigated in the ALEXA trial. In the crizotinib arm, 31.5% of the patients without BM at study entry developed BM during further course of the study, compared to only 4.6% of the patients in the alectinib group [10].
Furthermore, the addition of durvalumab to radiochemotherapy in stage III NSCLC was associated with an improved primary BM prevention as 5.5% of patients in the durvalumab arm developed BM compared to 11.0% in the control arm [28].

Conclusion

Remarkable intracranial efficacies were observed for several next-generation TKIs and immune checkpoint inhibitors—especially in patients with asymptomatic BM. Therefore, in case of available systemic treatment option with proven high intracranial efficacy, upfront systemic treatment can be evaluated in BM patients with asymptomatic disease.
Furthermore, based on the promising primary and secondary BM preventions in some new therapeutic agents, effective prevention of BM also needs to be addressed in future trials.
Take home messages
  • Targeted and immune directed therapy presented notable intracranial response rates especially in asymptomatic brain metastases.
  • Some immunotherapy and targeted therapy agents have shown primary and secondary BM prevention in clinical trials.

Conflict of interest

A.S. Berghoff has research support from Daiichi Sankyo (≤ 10,000 €), Roche (> 10,000 €) and honoraria for lectures, consultation or advisory board participation from Roche Bristol-Meyers Squibb, Merck, Daiichi Sankyo (all < 5000 €) as well as travel support from Roche, Amgen and AbbVie. A. Steindl declares that she has no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher’s Note

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

Unsere Produktempfehlungen

Abo für kostenpflichtige Inhalte

Literatur
1.
Zurück zum Zitat Steindl A, Kreminger J, Moor E, et al. Clinical characterization of a real-life cohort of 6001 patients with brain metastases from solid cancers treated between 1986–2020. Ann Oncol. 2020;31:S397.CrossRef Steindl A, Kreminger J, Moor E, et al. Clinical characterization of a real-life cohort of 6001 patients with brain metastases from solid cancers treated between 1986–2020. Ann Oncol. 2020;31:S397.CrossRef
3.
Zurück zum Zitat Soffietti R, Abacioglu U, Baumert B, et al. Diagnosis and treatment of brain metastases from solid tumors: Guidelines from the European Association of neuro-oncology (EANO). Neuro Oncol. 2017;19:162–74.CrossRef Soffietti R, Abacioglu U, Baumert B, et al. Diagnosis and treatment of brain metastases from solid tumors: Guidelines from the European Association of neuro-oncology (EANO). Neuro Oncol. 2017;19:162–74.CrossRef
7.
Zurück zum Zitat Reungwetwattana T, Nakagawa K, Cho BC, et al. CNS response to osimertinib versus standard epidermal growth factor receptor tyrosine kinase inhibitors in patients with untreated EGFR-mutated advanced non-small-cell lung cancer. J Clin Oncol. 2018;36:3290–7.CrossRef Reungwetwattana T, Nakagawa K, Cho BC, et al. CNS response to osimertinib versus standard epidermal growth factor receptor tyrosine kinase inhibitors in patients with untreated EGFR-mutated advanced non-small-cell lung cancer. J Clin Oncol. 2018;36:3290–7.CrossRef
8.
Zurück zum Zitat Schuler M, Wu Y‑L, Hirsh V, et al. First-line Afatinib versus chemotherapy in patients with non-small cell lung cancer and common epidermal growth factor receptor gene mutations and brain metastases. J Thorac Oncol. 2016;11:380–90.CrossRef Schuler M, Wu Y‑L, Hirsh V, et al. First-line Afatinib versus chemotherapy in patients with non-small cell lung cancer and common epidermal growth factor receptor gene mutations and brain metastases. J Thorac Oncol. 2016;11:380–90.CrossRef
12.
Zurück zum Zitat Solomon BJ, Besse B, Bauer TM, et al (2018) Lorlatinib in patients with ALK-positive non-small-cell lung cancer: results from a global phase 2 study. Lancet Oncol 19(12):1654–67CrossRef Solomon BJ, Besse B, Bauer TM, et al (2018) Lorlatinib in patients with ALK-positive non-small-cell lung cancer: results from a global phase 2 study. Lancet Oncol 19(12):1654–67CrossRef
23.
Zurück zum Zitat Tawbi HA, Forsyth PA, Algazi A, et al. Combined nivolumab and Ipilimumab in melanoma metastatic to the brain. N Engl J Med. 2018;379:722–30.CrossRef Tawbi HA, Forsyth PA, Algazi A, et al. Combined nivolumab and Ipilimumab in melanoma metastatic to the brain. N Engl J Med. 2018;379:722–30.CrossRef
27.
Zurück zum Zitat Ramalingam SS, Gray JE, Ohe Y, et al. Osimertinib vs comparator EGFR-TKI as first-line treatment for EGFRm advanced NSCLC (FLAURA): final overall survival analysis. Ann Oncol. 2019;30(suppl):v851–934. Ramalingam SS, Gray JE, Ohe Y, et al. Osimertinib vs comparator EGFR-TKI as first-line treatment for EGFRm advanced NSCLC (FLAURA): final overall survival analysis. Ann Oncol. 2019;30(suppl):v851–934.
Metadaten
Titel
Brain metastases: new systemic treatment approaches
verfasst von
Ariane Steindl, MD PhD
Anna Sophie Berghoff
Publikationsdatum
22.05.2021
Verlag
Springer Vienna
Erschienen in
memo - Magazine of European Medical Oncology / Ausgabe 2/2021
Print ISSN: 1865-5041
Elektronische ISSN: 1865-5076
DOI
https://doi.org/10.1007/s12254-021-00709-1

Weitere Artikel der Ausgabe 2/2021

memo - Magazine of European Medical Oncology 2/2021 Zur Ausgabe