Skip to main content

A rare non-gadolinium enhancing sarcoma brain metastasis with microenvironment dominated by tumor-associated macrophages

Abstract

Brain metastases occur in 1% of sarcoma cases and are associated with a median overall survival of 6 months. We report a rare case of a brain metastasis with unique radiologic and histopathologic features in a patient with low grade fibromyxoid sarcoma (LGFMS) previously treated with immune checkpoint inhibitor (ICI) therapy. The lone metastasis progressed in the midbrain tegmentum over 15 months as a non-enhancing, T2-hyperintense lesion with peripheral diffusion restriction, mimicking a demyelinating lesion. Histopathology of the lesion at autopsy revealed a rich infiltrate of tumor-associated macrophages (TAMs) with highest density at the leading edge of the metastasis, whereas there was a paucity of lymphocytes, suggestive of an immunologically cold environment. Given the important immunosuppressive and tumor-promoting functions of TAMs in gliomas and carcinoma/melanoma brain metastases, this unusual case provides an interesting example of a dense TAM infiltrate in a much rarer sarcoma brain metastasis.

Introduction

The development of brain metastases depends on supportive interactions between cancer cells and the surrounding brain tumor microenvironment (TME). Neurons, glia, and immune cells support cancer growth through diverse mechanisms including neuron-to-cancer cell synapses, astrocyte-to-cancer cell gap junctions, and maintenance of immunosuppression [2, 8, 16, 18]. TAMs, which include CNS-resident microglia and monocyte-derived macrophages (MDMs) recruited from the periphery, are a major component of the TME with important tumor-promoting and immunosuppressive functions. Whereas the glioma TME contains abundant microglia, the TME of carcinoma and melanoma brain metastases contains a greater proportion of MDMs as well as infiltrating lymphocytes and neutrophils [4, 9]. Sarcoma brain metastases are very rare, occurring in 1% of cases [5], and the TME of these brain metastases has not been well studied. We present a case of a sarcoma brain metastasis with unique imaging and histopathologic features, including high density of TAMs in the TME.

Case presentation

A 44-year-old woman with history of recurrent, metastatic LGFMS of the left piriformis muscle previously treated with ICIs presented with 1 month of gradually worsening binocular diagonal diplopia. Over the ten years prior to onset of neurologic symptoms, the LGFMS was treated with three surgical resections in the left gluteal region (pathology showed Fédération Nationale des Centres de Lutte Contre le Cancer grade 2 of 3, all resections had positive margins), two courses of radiation to the left pelvic region, and resection of a right cervical paraspinal metastasis followed by adjuvant radiation. Two years prior to presentation, there was progression of sarcoma in the pelvis as well as metastatic disease in the lungs, kidneys, and bone, with pathologic confirmation of bone involvement. She was started on a clinical trial in which she received IV ipilimumab 1 mg/kg and IV nivolumab 3 mg/kg (ICIs) every 3 weeks for four cycles, followed by nivolumab 480 mg every 4 weeks. Cryoablation of a left gluteal mass was performed after the first infusion of ipilimumab and nivolumab. ICI treatment was complicated by thyroiditis, adrenal insufficiency, and hepatitis that were managed with corticosteroids and hormone replacement. ICIs were discontinued after 9 months of therapy, 8 months prior to her presentation with neurologic symptoms. At the time of diplopia onset, there was a growing left adrenal metastasis being monitored on PET/CT. Neurologic exam showed right hypertropia worse with right and down gaze, and mild bilateral ptosis.

MRI of the brain revealed an 8 mm T2 hyperintense lesion in the right paramedian midbrain without restricted diffusion or enhancement. In retrospect, the midbrain lesion was subtly present on an MRI obtained 9 months prior (Fig. 1A, B). An extensive infectious and inflammatory workup including serum MOG-IgG and AQP-4 IgG antibodies and cerebrospinal fluid analysis including cytology, flow cytometry, oligoclonal bands, JC virus PCR, and autoimmune antibody panel was negative. MRI of the spine was negative for spinal cord lesions. The suspected diagnosis was CNS demyelination secondary to ICI, and she was treated with IV methylprednisolone 1 g for three days, followed by maintenance oral prednisone (30–60 mg daily), without improvement in her diplopia.

Fig. 1
figure 1

Radiologic progression of midbrain lesion. (A) Subtle T2/FLAIR-hyperintense lesion (red arrow) seen in retrospect 9 months prior to presentation with neurologic symptoms. (B) Right paramedian midbrain lesion at onset of neurologic symptoms, without contrast enhancement or diffusion restriction. (C-E) Growth of the lesion over 5 months, with gradual development of peripheral diffusion restriction (red arrows) but still no contrast enhancement. (F) FDG-PET shows midbrain lesion (red arrow) is hypometabolic compared to grey matter (*).

Three months after her presentation with diplopia, she developed fatigue accompanied by severe bilateral ptosis and ophthalmoplegia and left arm ataxia. MRI showed expansion of the non-enhancing T2-hyperintense midbrain lesion, now extending into the right medial thalamus and pons (Fig. 1C, D). The differential diagnosis was expanded to include glioma, however biopsy was deemed unsafe given the rostral brainstem location. Metastasis was not considered given the infiltrative and non-enhancing nature of the lesion. Further empiric treatment for an autoimmune etiology was given with IVIg 2 g/kg over three days. The patient underwent resection of the left adrenal mass, with pathology demonstrating metastatic sarcoma with high grade features including elevated mitotic rate of at least 30 mitotic figures per 10 high-power fields and 20% of the tumor with necrosis. The morphologic and immunophenotypic findings from the adrenal resection were interpreted as most consistent with sclerosing epithelioid fibrosarcoma (SEF), and the classic features of LGFMS were not seen. The high grade features consistent with SEF in the adrenal mass likely represented progression of the patient’s prior LGFMS.

Four months after presentation she developed worsening balance and falls and underwent five sessions of plasma exchange over six days. Five months after presentation she developed left-sided weakness, dysarthria and somnolence suggestive of involvement of the reticular activating system. MRI showed continued mild increase in the size of the midbrain lesion, with gradual development of peripheral diffusion restriction but still no contrast enhancement (Fig. 1E). PET/CT showed the midbrain lesion to be hypometabolic compared to grey matter (Fig. 1F), favoring an inflammatory rather than neoplastic process. She developed dysphagia requiring PEG tube placement and urinary retention requiring intermittent catheterization. Her clinical status deteriorated further over the next month with non-convulsive status epilepticus treated with levetiracetam and lacosamide. Despite resolution of seizures, her mental status remained depressed requiring invasive ventilation. She was transitioned to hospice care and died 6 months after presenting with neurologic symptoms.

Autopsy revealed metastatic sarcoma involving the midbrain, pons, and thalami (Fig. 2A). The sarcoma centered on the periaqueductal region with accompanying disseminated single-cell infiltration throughout the midbrain. The neoplastic cells stained positive for MUC-4 (Fig. 2A) and harbored a TAOK1::FUS rearrangement. While FUS fusions are common in some sarcoma subtypes, the TAOK1::FUS fusion has not been previously reported. The lesional midbrain and periaqueductal parenchyma were notable for extensive infiltration of CD68 + and CD163 + TAMs (Fig. 2B, C). The number of TAMs was particularly extensive relative to the number of tumor cells present, with highest TAM density at the leading edge of the metastasis, a location correlating with diffusion restriction on MRI (Fig. 2B, rightmost panel). In contrast to the abundant TAMs, there was a relative paucity of lymphocytes including CD3 + T cells and CD20 + B cells (Fig. 2D). Lymphocyte counts across 10 randomly sampled 1 mm2 regions revealed 17 ± 10 (mean ± standard deviation) CD3 + T cells per mm2 and 2 ± 1 CD20 + B cells per mm2. There was no evidence of tumor infiltrating the leptomeninges.

Fig. 2
figure 2

Histopathologic findings in the midbrain at autopsy. (A) Lesion contains MUC4 + sarcoma cells. (B) Extensive infiltration of CD68 + and CD163 + TAMs, with highest density at the leading edge of the metastasis corresponding to diffusion restriction on MRI (red arrows). (C) High magnification images of lesion depict morphology of CD68 + TAMs. Scale bar = 100 μm. (D) Immunohistochemical staining within the boxed region at left reveals a paucity of CD3 + T cells and CD20 + B cells compared to abundant CD163 + TAMs. Scale bar = 250 μm

Discussion

LGFMS has an incidence of 0.18 per million and typically presents in the extremities and trunk [13]. LGFMS may metastasize to the lungs and retroperitoneum [13], but there are no previous reports of brain metastases from systemic sites. There are six reports of primary intracranial LGFMS, all of which were well-circumscribed enhancing masses [1, 20]. SEF is a rare and aggressive soft-tissue sarcoma first described as a variant of LGFMS. LGFMS and SEF may appear together as a hybrid tumor, and they share diagnostic features including MUC4 immunoreactivity and FUS gene rearrangements [19]. SEF has distant metastatic spread in 40–80% of cases, and in a series of 88 patients with metastatic SEF, 2.9% had brain metastases [21]. This case represents an already rare sarcoma brain metastasis with additional unique and confounding radiologic and histopathologic features.

The sarcoma brain metastasis microenvironment featured extensive TAM infiltration and a dearth of lymphocytes. This contrasts with the relative abundance of lymphocytes found in the TME of more common brain metastases from breast, lung, and melanoma tumors [4, 9]. Interestingly, this case shared some characteristics with diffuse midline glioma (DMG), including the lack of enhancement and brainstem location, and DMG also has a non-inflammatory immune environment with abundance of TAMs, low levels of inflammatory cytokines, and a paucity of lymphocytes [10]. TAMs promote tumor growth by inducing immunosuppression, remodeling the extracellular matrix, and stimulating angiogenesis [16, 17, 22]. In vitro models of brain metastasis show microglia attaching themselves to tumor cells and actively transporting them into brain tissue [15]. TAMs have an inhibitory effect on T cell recruitment and function [3], which may have contributed to the paucity of lymphocytes and weak immune response in this case. Even if this patient had been eligible for further ICIs, the lack of lymphocytes suggests the lesion would have responded poorly to immunotherapy [11].

Peripheral diffusion restriction is rare for brain metastases and is more commonly seen in central nervous system demyelinating lesions, where it is attributed to clustering of activated microglia [6]. The peripheral diffusion restriction in this case coincided with dense infiltration of TAMs at the leading edge of the brain metastasis, implying that tightly packed TAMs restricted water diffusion akin to demyelinating lesions. Additionally, the persistent lack of contrast enhancement is unusual for brain metastases, which are typically fueled by vascular endothelial growth factor (VEGF)-dependent angiogenesis associated with a disrupted blood-brain barrier. Non-enhancement of brain metastases may occur after receipt of the anti-VEGF antibody bevacizumab [7], which this patient did not receive. Thus, the absence of contrast enhancement in this case suggests that VEGF-mediated angiogenesis was not required for metastasis growth. Interestingly, TAMs mediate resistance to antiangiogenic brain tumor therapies in preclinical models, and TAMs are increased at autopsy in recurrent glioblastoma patients who received antiangiogenic therapies compared with patients who did not [12, 14]. Rebound vascularization does not occur in TAM-mediated antiangiogenic resistance, underscoring that TAMs support cancer cells in diverse ways such as promoting tumor cell migration, invasion, and immunosuppression [12]. This case with extensive TAMs and an intact blood-brain barrier is consistent with prior studies showing that TAMs facilitate brain tumor growth in many ways beyond angiogenesis.

In sum, we report an extremely rare fibromyxoid sarcoma brain metastasis with unique radiologic and histopathologic features, highlighting the role of the TME and showing that non-enhancing lesions may represent brain metastases in certain contexts. The dense infiltration of TAMs together with scarce lymphocytes seen in this case raises the question whether TAMs might have important immunosuppressive and tumor-promoting roles in sarcoma brain metastases, similar to other more common brain tumors. Future research will determine if therapies that alter TAM phenotypes to create a hostile TME are effective as brain tumor treatments.

Data availability

All data generated or analyzed during this study are included in this published article.

Abbreviations

TAM:

Tumor-associated macrophages

ICI:

Immune checkpoint inhibitor

TME:

Tumor microenvironment

VEGF:

Vascular endothelial growth factor

LGFMS:

Low grade fibromyxoid sarcoma

MDM:

Monocyte-derived macrophages

SEF:

Sclerosing epithelioid fibrosarcoma

TIL:

Tumor-infiltrating lymphocyte

References

  1. Chen N, Gong J, Nie L, Chen X, Xu M, Chen M et al (2015) Primary intracranial low-grade fibromyxoid sarcoma with FUS gene rearrangement. Neuropathology 35:348–353. https://doi.org/10.1111/neup.12197

    Article  CAS  PubMed  Google Scholar 

  2. Chen Q, Boire A, Jin X, Valiente M, Er EE, Lopez-Soto A et al (2016) Carcinoma–astrocyte gap junctions promote brain Metastasis by cGAMP transfer. Nature 533:493–498. https://doi.org/10.1038/nature18268

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. DeNardo DG, Ruffell B (2019) Macrophages as regulators of tumour immunity and immunotherapy. Nat Rev Immunol 19:369–382. https://doi.org/10.1038/s41577-019-0127-6

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Friebel E, Kapolou K, Unger S, Núñez NG, Utz S, Rushing EJ et al (2020) Single-cell mapping of Human Brain Cancer reveals tumor-specific instruction of tissue-invading leukocytes. Cell 181:1626–1642e1620. https://doi.org/10.1016/j.cell.2020.04.055

    Article  CAS  PubMed  Google Scholar 

  5. Gusho CA, Blank AT, Batus M (2021) Outcomes of brain Metastasis in high-grade bone and soft tissue sarcoma: an analysis of clinicopathological characteristics and survival data. Rare Tumors 13:20363613211026151. https://doi.org/10.1177/20363613211026151

    Article  PubMed  PubMed Central  Google Scholar 

  6. Hyland M, Bermel RA, Cohen JA (2013) Restricted diffusion preceding gadolinium enhancement in large or tumefactive demyelinating lesions. Neurol Clin Pract 3:15–21. https://doi.org/10.1212/CPJ.0b013e318283ff8e

    Article  PubMed  PubMed Central  Google Scholar 

  7. Karimi S, Lis E, Gilani S, D’Ambrosio N, Holodny A (2011) Nonenhancing brain metastases. J Neuroimaging 21:184–187. https://doi.org/10.1111/j.1552-6569.2009.00389.x

    Article  PubMed  Google Scholar 

  8. Khan F, Pang L, Dunterman M, Lesniak MS, Heimberger AB, Chen P (2023) Macrophages and microglia in glioblastoma: heterogeneity, plasticity, and therapy. J Clin Investig 133. https://doi.org/10.1172/JCI163446

  9. Klemm F, Maas RR, Bowman RL, Kornete M, Soukup K, Nassiri S et al (2020) Interrogation of the Microenvironmental Landscape in Brain Tumors reveals Disease-specific alterations of Immune cells. Cell 181:1643–1660e1617. https://doi.org/10.1016/j.cell.2020.05.007

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Lin GL, Nagaraja S, Filbin MG, Suvà ML, Vogel H, Monje M (2018) Non-inflammatory Tumor microenvironment of diffuse intrinsic pontine glioma. 6 51. https://doi.org/10.1186/s40478-018-0553-x

  11. Linette GP, Carreno BM (2019) Tumor-infiltrating lymphocytes in the checkpoint inhibitor era. Curr Hematol Malig Rep 14:286–291. https://doi.org/10.1007/s11899-019-00523-x

    Article  PubMed  PubMed Central  Google Scholar 

  12. Lu-Emerson C, Snuderl M, Kirkpatrick ND, Goveia J, Davidson C, Huang Y et al (2013) Increase in tumor-associated macrophages after antiangiogenic therapy is associated with poor survival among patients with recurrent glioblastoma. Neurooncology 15:1079–1087. https://doi.org/10.1093/neuonc/not082

    Article  CAS  Google Scholar 

  13. Maretty-Nielsen K, Baerentzen S, Keller J, Dyrop HB, Safwat A (2013) Low-Grade Fibromyxoid Sarcoma: incidence, Treatment Strategy of Metastases, and clinical significance of the FUS Gene. Sarcoma 2013:256280. https://doi.org/10.1155/2013/256280

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Piao Y, Liang J, Holmes L, Zurita AJ, Henry V, Heymach JV et al (2012) Glioblastoma resistance to anti-VEGF therapy is associated with myeloid cell infiltration, stem cell accumulation, and a mesenchymal phenotype. Neurooncology 14:1379–1392. https://doi.org/10.1093/neuonc/nos158

    Article  CAS  Google Scholar 

  15. Pukrop T, Dehghani F, Chuang HN, Lohaus R, Bayanga K, Heermann S et al (2010) Microglia promote colonization of brain tissue by Breast cancer cells in a wnt-dependent way. Glia 58:1477–1489. https://doi.org/10.1002/glia.21022

    Article  PubMed  Google Scholar 

  16. Schulz M, Sevenich L (2021) TAMs in Brain Metastasis: Molecular signatures in Mouse and Man. Front Immunol 12:716504. https://doi.org/10.3389/fimmu.2021.716504

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Sevenich L, Bowman RL, Mason SD, Quail DF, Rapaport F, Elie BT et al (2014) Analysis of tumour- and stroma-supplied proteolytic networks reveals a brain-metastasis-promoting role for cathepsin S. Nat Cell Biol 16:876–888. https://doi.org/10.1038/ncb3011

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Solomiia S, Kaylee G, Wengang W, Elana C, Belgin Y, Yin L et al (2023) Neuronal-activity dependent mechanisms of small cell Lung Cancer Progression. bioRxiv: 2023.2001.2019.524430. https://doi.org/10.1101/2023.01.19.524430

  19. Warmke LM, Meis JM (2021) Sclerosing Epithelioid Fibrosarcoma: a distinct sarcoma with aggressive features. Am J Surg Pathol 45:317–328. https://doi.org/10.1097/pas.0000000000001559

    Article  PubMed  Google Scholar 

  20. White IK, Scherer AG, Baumanis MM, Abdulkader M, Fulkerson DH (2015) Rapidly enlarging low-grade fibromyxoid sarcoma with intracranial extension in a 5-year-old girl: case report. J Neurosurg Pediatr 16:372–376. https://doi.org/10.3171/2015.3.peds14564

    Article  PubMed  Google Scholar 

  21. Woods AD, Purohit R, Mitchell LC, Collier JR, Collier KA, Lathara M et al (2021) Metastatic pediatric sclerosing epithelioid fibrosarcoma. 7. https://doi.org/10.1101/mcs.a006093

  22. Zhu C, Kros JM, Cheng C, Mustafa D (2017) The contribution of tumor-associated macrophages in glioma neo-angiogenesis and implications for anti-angiogenic strategies. Neurooncology 19:1435–1446. https://doi.org/10.1093/neuonc/nox081

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

No funding sources applicable.

Author information

Authors and Affiliations

Authors

Contributions

DR analyzed and interpreted the clinical, radiologic, and pathologic data, and prepared draft of the manuscript. JW performed the histological examination of the brain. EN, WZ, EV, GC, QM, and KG made substantial contributions to interpretation of patient data. NF and MI interpreted radiology images. HV interpreted and supervised histopathologic data interpretation. SN interpreted data and supervised the project. All authors read, revised, and approved the final manuscript.

Corresponding author

Correspondence to David Rogawski.

Ethics declarations

Ethics approval and consent to participate

Consent was obtained from the patient’s next of kin.

Consent for publication

Consent was obtained from the patient’s next of kin.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

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

Rights and permissions

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/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rogawski, D., Wheeler, J., Nie, E. et al. A rare non-gadolinium enhancing sarcoma brain metastasis with microenvironment dominated by tumor-associated macrophages. acta neuropathol commun 12, 15 (2024). https://doi.org/10.1186/s40478-023-01713-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40478-023-01713-8

Keywords