Pub Date : 2025-12-19DOI: 10.1186/s40478-025-02180-z
Szymon Baluszek, Paulina Kober, Michał Wa̧grodzki, Jacek Kunicki, Bartosz Wojtaś, Paulina Szadkowska, Bożena Kamińska, Thibault Passeri, Tomasz Mandat, Mateusz Bujko
Introduction: Dedifferentiated (DC) and poorly differentiated chordomas (PDC) are rare, aggressive chordomas with a significantly worse prognosis than conventional chordomas (CC). The molecular mechanisms driving them remain poorly understood.
Methods: Matched primary CC and recurrent DC cryopreserved samples from one patient were analyzed with whole-exome sequencing (WES). Samples from three additional DCs and one PDC underwent targeted sequencing of cancer-related genes. Furthermore, 102 CC cases - 32 novel and 70 from literature, were analyzed. Functional and survival analysis was performed.
Results: WES revealed striking genomic changes during progression from CC to DC, with the number of somatic mutations increasing from 211 in primary to 430 in the recurrent DC; recurrence acquired TP53 and BRCA1 deleterious mutations, along with copy-number alterations, including loss of 6q containing the TBXT locus. Targeted sequencing identified TP53 mutations in 4/5 DC&PDC cases compared to 1/102 cases in combined CC cohorts (p = 2.7×10-5, OR=162.9). In 3 recurrent DC samples with TP53 variant, presence of the mutation was assessed in primary CC sample and in neither, this variant was found. Literature review revealed TP53 mutations in 9/23 (39%) DC&PDC cases versus 5/445 (1.24%) CC cases. Survival analysis demonstrated that TP53 mutations confer a significantly worse prognosis in DC patients (p = 0.03).
Conclusion: TP53 mutations are acquired during chordoma progression and are associated with an aggressive phenotype; TP53 sequencing could serve as a prognostic and potentially predictive biomarker in aggressive chordomas.
{"title":"TP53 mutations as drivers of chordoma progression and hallmarks of aggressive chordoma.","authors":"Szymon Baluszek, Paulina Kober, Michał Wa̧grodzki, Jacek Kunicki, Bartosz Wojtaś, Paulina Szadkowska, Bożena Kamińska, Thibault Passeri, Tomasz Mandat, Mateusz Bujko","doi":"10.1186/s40478-025-02180-z","DOIUrl":"https://doi.org/10.1186/s40478-025-02180-z","url":null,"abstract":"<p><strong>Introduction: </strong>Dedifferentiated (DC) and poorly differentiated chordomas (PDC) are rare, aggressive chordomas with a significantly worse prognosis than conventional chordomas (CC). The molecular mechanisms driving them remain poorly understood.</p><p><strong>Methods: </strong>Matched primary CC and recurrent DC cryopreserved samples from one patient were analyzed with whole-exome sequencing (WES). Samples from three additional DCs and one PDC underwent targeted sequencing of cancer-related genes. Furthermore, 102 CC cases - 32 novel and 70 from literature, were analyzed. Functional and survival analysis was performed.</p><p><strong>Results: </strong>WES revealed striking genomic changes during progression from CC to DC, with the number of somatic mutations increasing from 211 in primary to 430 in the recurrent DC; recurrence acquired TP53 and BRCA1 deleterious mutations, along with copy-number alterations, including loss of 6q containing the TBXT locus. Targeted sequencing identified TP53 mutations in 4/5 DC&PDC cases compared to 1/102 cases in combined CC cohorts (p = 2.7×10<sup>-5</sup>, OR=162.9). In 3 recurrent DC samples with TP53 variant, presence of the mutation was assessed in primary CC sample and in neither, this variant was found. Literature review revealed TP53 mutations in 9/23 (39%) DC&PDC cases versus 5/445 (1.24%) CC cases. Survival analysis demonstrated that TP53 mutations confer a significantly worse prognosis in DC patients (p = 0.03).</p><p><strong>Conclusion: </strong>TP53 mutations are acquired during chordoma progression and are associated with an aggressive phenotype; TP53 sequencing could serve as a prognostic and potentially predictive biomarker in aggressive chordomas.</p>","PeriodicalId":6914,"journal":{"name":"Acta Neuropathologica Communications","volume":" ","pages":""},"PeriodicalIF":5.7,"publicationDate":"2025-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145792996","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-19DOI: 10.1186/s40478-025-02171-0
Jonas Yeung, Prisca Hsu, Jordan Mak, Ali Darbandi, Anne L Wheeler, Rosanna Weksberg, Sharon L Guger, Russell J Schachar, Shinya Ito, Johann Hitzler, Brian J Nieman
Vincristine is an essential chemotherapy agent administered for various pediatric cancers including acute lymphoblastic leukemia (ALL). While multi-agent chemotherapy for pediatric ALL is highly curative, with survival approaching 95%, it carries the risk of irreversible neurocognitive late effects. Vincristine is known to cause peripheral neuropathy, but its relationship to brain toxicity remains understudied. We investigated vincristine-mediated brain toxicity in young mice lacking Sarm1, a gene whose deletion protects against vincristine-induced peripheral neuropathy. Littermate wildtype and knockout mice were randomly assigned to saline or vincristine groups. In vivo MRI was performed from childhood to early adulthood to measure brain structure volumes, followed by ex vivo diffusion tensor imaging (DTI) to assess microstructural changes. In a separate cohort, electron microscopy (EM) quantified axon morphology in the sciatic nerve and corpus callosum. Vincristine induced significant volume reduction across the brain, while Sarm1 knockout reduced loss in both grey and white matter. Several regions, including the amygdala and dentate gyrus, showed near-complete recovery in knockouts. DTI revealed limited changes with no genotype differences. EM demonstrated vincristine-induced axon morphology alterations in wildtype mice in both the sciatic nerve and corpus callosum. Sarm1 knockout rescued sciatic nerve morphology but not corpus callosum axons. These findings suggest that SARM1-mediated peripheral axon damage may contribute to vincristine-induced brain volume deficits, whereas brain axons may be affected through distinct, SARM1-independent mechanisms. These results suggest a link between vincristine-induced peripheral axon damage and alterations in brain development, with implications for neurocognitive deficits experienced by ALL survivors. Our results suggest that mitigating vincristine-induced peripheral neuropathy may also help reduce neurocognitive deficits in pediatric patients undergoing vincristine treatment.
{"title":"Vincristine-induced brain toxicity is reduced with prevention of peripheral axon degeneration in Sarm1 knockout mice.","authors":"Jonas Yeung, Prisca Hsu, Jordan Mak, Ali Darbandi, Anne L Wheeler, Rosanna Weksberg, Sharon L Guger, Russell J Schachar, Shinya Ito, Johann Hitzler, Brian J Nieman","doi":"10.1186/s40478-025-02171-0","DOIUrl":"10.1186/s40478-025-02171-0","url":null,"abstract":"<p><p>Vincristine is an essential chemotherapy agent administered for various pediatric cancers including acute lymphoblastic leukemia (ALL). While multi-agent chemotherapy for pediatric ALL is highly curative, with survival approaching 95%, it carries the risk of irreversible neurocognitive late effects. Vincristine is known to cause peripheral neuropathy, but its relationship to brain toxicity remains understudied. We investigated vincristine-mediated brain toxicity in young mice lacking Sarm1, a gene whose deletion protects against vincristine-induced peripheral neuropathy. Littermate wildtype and knockout mice were randomly assigned to saline or vincristine groups. In vivo MRI was performed from childhood to early adulthood to measure brain structure volumes, followed by ex vivo diffusion tensor imaging (DTI) to assess microstructural changes. In a separate cohort, electron microscopy (EM) quantified axon morphology in the sciatic nerve and corpus callosum. Vincristine induced significant volume reduction across the brain, while Sarm1 knockout reduced loss in both grey and white matter. Several regions, including the amygdala and dentate gyrus, showed near-complete recovery in knockouts. DTI revealed limited changes with no genotype differences. EM demonstrated vincristine-induced axon morphology alterations in wildtype mice in both the sciatic nerve and corpus callosum. Sarm1 knockout rescued sciatic nerve morphology but not corpus callosum axons. These findings suggest that SARM1-mediated peripheral axon damage may contribute to vincristine-induced brain volume deficits, whereas brain axons may be affected through distinct, SARM1-independent mechanisms. These results suggest a link between vincristine-induced peripheral axon damage and alterations in brain development, with implications for neurocognitive deficits experienced by ALL survivors. Our results suggest that mitigating vincristine-induced peripheral neuropathy may also help reduce neurocognitive deficits in pediatric patients undergoing vincristine treatment.</p>","PeriodicalId":6914,"journal":{"name":"Acta Neuropathologica Communications","volume":"13 1","pages":"254"},"PeriodicalIF":5.7,"publicationDate":"2025-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12717744/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145792915","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-18DOI: 10.1186/s40478-025-02204-8
Emma Noël, Fabien Guimiot, Yline Capri, Marianne Alison, Asha Baskaran, Clémence Delcour, David Germanaud, Sophie Lebon, Caroline Storey, Nicolas de Roux, Adeline Orts-Del'Immagine
{"title":"Biallelic null RAB3GAP1 variants impair cortical development and autophagy in Warburg Micro syndrome: evidence from fetal brain tissue and patient fibroblasts.","authors":"Emma Noël, Fabien Guimiot, Yline Capri, Marianne Alison, Asha Baskaran, Clémence Delcour, David Germanaud, Sophie Lebon, Caroline Storey, Nicolas de Roux, Adeline Orts-Del'Immagine","doi":"10.1186/s40478-025-02204-8","DOIUrl":"https://doi.org/10.1186/s40478-025-02204-8","url":null,"abstract":"","PeriodicalId":6914,"journal":{"name":"Acta Neuropathologica Communications","volume":" ","pages":""},"PeriodicalIF":5.7,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145779922","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-17DOI: 10.1186/s40478-025-02189-4
Alexia Tiberi, Elena Montagni, Giulia Borgonovo, Eléa Coulomb, Laura Restani, Anna Letizia Allegra Mascaro, Simona Capsoni, Antonino Cattaneo
{"title":"Microglia drive synaptic and functional connectivity deficits in the Ts65Dn mouse model of Down syndrome by affecting inhibition.","authors":"Alexia Tiberi, Elena Montagni, Giulia Borgonovo, Eléa Coulomb, Laura Restani, Anna Letizia Allegra Mascaro, Simona Capsoni, Antonino Cattaneo","doi":"10.1186/s40478-025-02189-4","DOIUrl":"https://doi.org/10.1186/s40478-025-02189-4","url":null,"abstract":"","PeriodicalId":6914,"journal":{"name":"Acta Neuropathologica Communications","volume":" ","pages":""},"PeriodicalIF":5.7,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145773184","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-17DOI: 10.1186/s40478-025-02173-y
Ashley R Tetens, Tyler R Findlay, Jordyn Craig-Schwartz, Athanasia Liapodimitri, Oscar Camacho, Kegan O Skalitzky, Adrian Idrizi, Rakel Tryggvadottir, Kayleigh Lunsford, Eric H Raabe, Michael A Koldobskiy
{"title":"Disordered DNA methylation leads to targetable transcriptional plasticity in ATRT.","authors":"Ashley R Tetens, Tyler R Findlay, Jordyn Craig-Schwartz, Athanasia Liapodimitri, Oscar Camacho, Kegan O Skalitzky, Adrian Idrizi, Rakel Tryggvadottir, Kayleigh Lunsford, Eric H Raabe, Michael A Koldobskiy","doi":"10.1186/s40478-025-02173-y","DOIUrl":"https://doi.org/10.1186/s40478-025-02173-y","url":null,"abstract":"","PeriodicalId":6914,"journal":{"name":"Acta Neuropathologica Communications","volume":" ","pages":""},"PeriodicalIF":5.7,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145773225","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-16DOI: 10.1186/s40478-025-02138-1
Caroline Liénard, Nicolas Pradeilles, Elisabeth Cortier, Cedric Hassen-Khodja, Leticia Arias, Maria Ceprian-Costoso, Antoine Picot, Anne-Laure Mausset-Bonnefont, Chantal Cazevieille, Frederic Fiore, Pascale Bomont
The nervous system evolved a variety of connections and neuron types to sustain diverse functions. While challenging, unlocking the universal mechanisms that support neuron integrity can be addressed in giant axonal neuropathy (GAN), a rare and fatal disease with broad deterioration of the nervous system. Here, we describe a new mouse strain that recapitulates key aspects of the GAN pathology following the introduction of a disease-causing mutation in GAN. Unlike previous GAN knock-out mice which show no overt phenotype, GANA49E/A49E mice exhibit early sensory-motor deficits and ataxia, giant axons and demyelination which, together with increased abundance, dramatic compaction and disorganization of neurofilaments across the nervous system, mimics the human disease. Using this model, we uncover novel alterations within neuromuscular junctions and muscles that might contribute to GAN pathogenesis. Interestingly, we pinpoint a sex bias whereby females show more severe histopathological damage and disease severity. Altogether, the GANA49E strain provides the first robust rodent model for GAN, recapitulating the symptoms and histological hallmarks of the human pathology. This model will be invaluable when investigating the cellular and molecular mechanisms that uphold neuron integrity along with effective therapies for GAN.
{"title":"Disease mutation in gigaxonin-E3 ligase recapitulates giant axonal neuropathy in mice.","authors":"Caroline Liénard, Nicolas Pradeilles, Elisabeth Cortier, Cedric Hassen-Khodja, Leticia Arias, Maria Ceprian-Costoso, Antoine Picot, Anne-Laure Mausset-Bonnefont, Chantal Cazevieille, Frederic Fiore, Pascale Bomont","doi":"10.1186/s40478-025-02138-1","DOIUrl":"https://doi.org/10.1186/s40478-025-02138-1","url":null,"abstract":"<p><p>The nervous system evolved a variety of connections and neuron types to sustain diverse functions. While challenging, unlocking the universal mechanisms that support neuron integrity can be addressed in giant axonal neuropathy (GAN), a rare and fatal disease with broad deterioration of the nervous system. Here, we describe a new mouse strain that recapitulates key aspects of the GAN pathology following the introduction of a disease-causing mutation in GAN. Unlike previous GAN knock-out mice which show no overt phenotype, GAN<sup>A49E/A49E</sup> mice exhibit early sensory-motor deficits and ataxia, giant axons and demyelination which, together with increased abundance, dramatic compaction and disorganization of neurofilaments across the nervous system, mimics the human disease. Using this model, we uncover novel alterations within neuromuscular junctions and muscles that might contribute to GAN pathogenesis. Interestingly, we pinpoint a sex bias whereby females show more severe histopathological damage and disease severity. Altogether, the GAN<sup>A49E</sup> strain provides the first robust rodent model for GAN, recapitulating the symptoms and histological hallmarks of the human pathology. This model will be invaluable when investigating the cellular and molecular mechanisms that uphold neuron integrity along with effective therapies for GAN.</p>","PeriodicalId":6914,"journal":{"name":"Acta Neuropathologica Communications","volume":" ","pages":""},"PeriodicalIF":5.7,"publicationDate":"2025-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145766824","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-13DOI: 10.1186/s40478-025-02203-9
Liyun Jia, Yue Chen, Hengzeng Li, Kai Zhao, Shuo Ge, Cong Wang, Jiayuan Zhao, Feifei Li, Leilei Zhang, Anhui Yao
Dysfunction of the glymphatic system (GS), a brain-wide waste clearance pathway dependent on polarized aquaporin-4 (AQP4) water channels on astrocytic endfeet, is increasingly recognized as a critical mechanism in both neurodegenerative diseases and brain tumors. In Alzheimer's (AD) and Parkinson's (PD) diseases, impaired glymphatic function leads to the accumulation of neurotoxic proteins, including amyloid-β (Aβ), tau, and α-synuclein (α-syn). Contributing factors include loss of AQP4 polarization, reduced arterial pulsatility, genetic risks (e.g., APOE4, FAM171A2 mutations), and sleep disturbances. These functional impairments can be quantified using neuroimaging biomarkers such as the diffusion tensor imaging along the perivascular space (DTI-ALPS) index and choroid plexus volume (CPV), which correlate with pathological burden and clinical decline, though the direct physiological interpretation of these metrics requires further validation. Conversely, in glioblastoma and other brain tumors, mechanical compression and lactate-driven acidosis obstruct perivascular fluid transport, promoting an immunosuppressive tumor microenvironment that limits T-cell infiltration and confers therapeutic resistance. Here, too, glymphatic dysfunction is reflected by a reduced ALPS index, which correlates with tumor grade, peritumoral edema, and survival. Emerging therapeutic strategies aimed at restoring GS function include pharmacological interventions (e.g., circadian regulators, AQP4 modulators), non-invasive techniques (e.g., cervical lymphatic stimulation, gamma stimulation, exercise), and surgical approaches (e.g., lymphatic-venous anastomosis). Advances in multimodal MRI and artificial intelligence (AI)-enhanced analytics further support novel diagnostic capabilities. This review highlights the dual role of the GS across neurological disorders and underscores its potential as a therapeutic target for enhancing waste clearance and immune modulation. However, significant challenges remain, including the validation of human biomarkers, elucidating bidirectional tumor-glymphatic crosstalk, and translating preclinical discoveries into clinical practice.
{"title":"The glymphatic system in neurodegenerative diseases and brain tumors: mechanistic insights, biomarker advances, and therapeutic opportunities.","authors":"Liyun Jia, Yue Chen, Hengzeng Li, Kai Zhao, Shuo Ge, Cong Wang, Jiayuan Zhao, Feifei Li, Leilei Zhang, Anhui Yao","doi":"10.1186/s40478-025-02203-9","DOIUrl":"https://doi.org/10.1186/s40478-025-02203-9","url":null,"abstract":"<p><p>Dysfunction of the glymphatic system (GS), a brain-wide waste clearance pathway dependent on polarized aquaporin-4 (AQP4) water channels on astrocytic endfeet, is increasingly recognized as a critical mechanism in both neurodegenerative diseases and brain tumors. In Alzheimer's (AD) and Parkinson's (PD) diseases, impaired glymphatic function leads to the accumulation of neurotoxic proteins, including amyloid-β (Aβ), tau, and α-synuclein (α-syn). Contributing factors include loss of AQP4 polarization, reduced arterial pulsatility, genetic risks (e.g., APOE4, FAM171A2 mutations), and sleep disturbances. These functional impairments can be quantified using neuroimaging biomarkers such as the diffusion tensor imaging along the perivascular space (DTI-ALPS) index and choroid plexus volume (CPV), which correlate with pathological burden and clinical decline, though the direct physiological interpretation of these metrics requires further validation. Conversely, in glioblastoma and other brain tumors, mechanical compression and lactate-driven acidosis obstruct perivascular fluid transport, promoting an immunosuppressive tumor microenvironment that limits T-cell infiltration and confers therapeutic resistance. Here, too, glymphatic dysfunction is reflected by a reduced ALPS index, which correlates with tumor grade, peritumoral edema, and survival. Emerging therapeutic strategies aimed at restoring GS function include pharmacological interventions (e.g., circadian regulators, AQP4 modulators), non-invasive techniques (e.g., cervical lymphatic stimulation, gamma stimulation, exercise), and surgical approaches (e.g., lymphatic-venous anastomosis). Advances in multimodal MRI and artificial intelligence (AI)-enhanced analytics further support novel diagnostic capabilities. This review highlights the dual role of the GS across neurological disorders and underscores its potential as a therapeutic target for enhancing waste clearance and immune modulation. However, significant challenges remain, including the validation of human biomarkers, elucidating bidirectional tumor-glymphatic crosstalk, and translating preclinical discoveries into clinical practice.</p>","PeriodicalId":6914,"journal":{"name":"Acta Neuropathologica Communications","volume":" ","pages":""},"PeriodicalIF":5.7,"publicationDate":"2025-12-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145751440","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-11DOI: 10.1186/s40478-025-02202-w
Johannes Wach, Alim Emre Basaran, Obada T Alhalabi, Jürgen Beck, Vicki M Butenschoen, Steven D Chang, Marcus Czabanka, Tomasz Czernicki, Philipp Dammann, Roberto Doria-Medina, Sven Oliver Eicker, Alonso Barrantes-Freer, Christine Gizaw, Erdem Güresir, Marc Hohenhaus, Romina Hohenhaus, Ahed H Kattaa, Fatma Kilinc, Lukas Klein, Nikolaus Kögl, Sandro Krieg, Przemyslaw Kunert, Maximilian Middelkamp, Bernhard Meyer, Nicolas Neidert, Julia Onken, Tobias Pantel, David J Park, Laurèl Rauschenbach, Roman Sankowski, Alejandro N Santos, Nils Ole Schmidt, Sebastian Siller, Ulrich Sure, Claudius Thomé, Tarik Tihan, Martin Vychopen, Peter Vajkoczy, Maria Wostrack, Jan-Helge Klingler
Spinal hemangioblastomas (sHB) are rare vascular tumors, with distinct clinical courses between von Hippel-Lindau (VHL)-associated and sporadic cases. The MIB-1 labeling index has been proposed as a surrogate marker for tumor proliferation, but its prognostic value remains unclear in this context. In this subgroup analysis from a multicenter retrospective study, we analyzed 116 primary sHB patients with available MIB-1 indices. Patients were stratified by VHL status. Statistical comparisons included ROC analyses for local progression-free survival (PFS) prediction and Kaplan-Meier survival curves for local PFS, stratified by a MIB-1 index cut-off derived from Youden's index. The MIB-1 index was significantly lower in VHL-associated tumors compared to sporadic ones (mean 2.17% vs. 3.02%, p = 0.008). In VHL-associated sHB, a higher MIB-1 index (≥ 2%) correlated with an increased risk of local tumor progression (AUC 0.74, 95% CI 0.49-0.98), whereas this was not observed in sporadic cases (AUC 0.56, 95% CI 0.23-0.88). Kaplan-Meier analysis showed that VHL patients with MIB-1 ≥ 2% had significantly shorter PFS (p = 0.05), while no significant association was found in sporadic tumors (p = 0.87). Our findings suggest that while VHL-associated sHB exhibit lower proliferative indices overall, elevated MIB-1 labeling indices might serve as a prognostic marker of shorter local PFS in this subgroup. In contrast, MIB-1 index appears to have limited prognostic relevance in sporadic sHB. These results highlight the importance of further molecular stratification and proliferation assessment in sHB to better inform clinical decision-making.
脊髓血管母细胞瘤(sHB)是一种罕见的血管肿瘤,在von Hippel-Lindau (VHL)相关病例和散发性病例之间具有明显的临床病程。MIB-1标记指数已被提出作为肿瘤增殖的替代标记物,但其预后价值在这方面尚不清楚。在这个来自多中心回顾性研究的亚组分析中,我们分析了116例具有可用MIB-1指数的原发性sHB患者。根据VHL状态对患者进行分层。统计比较包括局部无进展生存期(PFS)预测的ROC分析和局部PFS的Kaplan-Meier生存曲线,通过来自约登指数的MIB-1指数分界进行分层。vhl相关肿瘤的mb -1指数明显低于散发性肿瘤(平均2.17%比3.02%,p = 0.008)。在vhl相关sHB中,较高的MIB-1指数(≥2%)与局部肿瘤进展风险增加相关(AUC 0.74, 95% CI 0.49-0.98),而在散发病例中未观察到这一点(AUC 0.56, 95% CI 0.23-0.88)。Kaplan-Meier分析显示,MIB-1≥2%的VHL患者PFS显著缩短(p = 0.05),而散发性肿瘤无显著相关性(p = 0.87)。我们的研究结果表明,虽然vhl相关sHB总体上表现出较低的增殖指数,但升高的MIB-1标记指数可能作为该亚组局部PFS较短的预后标志物。相反,在散发性sHB中,mb -1指数的预后相关性似乎有限。这些结果强调了sHB进一步分子分层和增殖评估的重要性,以更好地为临床决策提供信息。
{"title":"Prognostic relevance of MIB-1 labeling index in VHL-associated and sporadic spinal hemangioblastomas: a subgroup analysis from a multicentric study.","authors":"Johannes Wach, Alim Emre Basaran, Obada T Alhalabi, Jürgen Beck, Vicki M Butenschoen, Steven D Chang, Marcus Czabanka, Tomasz Czernicki, Philipp Dammann, Roberto Doria-Medina, Sven Oliver Eicker, Alonso Barrantes-Freer, Christine Gizaw, Erdem Güresir, Marc Hohenhaus, Romina Hohenhaus, Ahed H Kattaa, Fatma Kilinc, Lukas Klein, Nikolaus Kögl, Sandro Krieg, Przemyslaw Kunert, Maximilian Middelkamp, Bernhard Meyer, Nicolas Neidert, Julia Onken, Tobias Pantel, David J Park, Laurèl Rauschenbach, Roman Sankowski, Alejandro N Santos, Nils Ole Schmidt, Sebastian Siller, Ulrich Sure, Claudius Thomé, Tarik Tihan, Martin Vychopen, Peter Vajkoczy, Maria Wostrack, Jan-Helge Klingler","doi":"10.1186/s40478-025-02202-w","DOIUrl":"10.1186/s40478-025-02202-w","url":null,"abstract":"<p><p>Spinal hemangioblastomas (sHB) are rare vascular tumors, with distinct clinical courses between von Hippel-Lindau (VHL)-associated and sporadic cases. The MIB-1 labeling index has been proposed as a surrogate marker for tumor proliferation, but its prognostic value remains unclear in this context. In this subgroup analysis from a multicenter retrospective study, we analyzed 116 primary sHB patients with available MIB-1 indices. Patients were stratified by VHL status. Statistical comparisons included ROC analyses for local progression-free survival (PFS) prediction and Kaplan-Meier survival curves for local PFS, stratified by a MIB-1 index cut-off derived from Youden's index. The MIB-1 index was significantly lower in VHL-associated tumors compared to sporadic ones (mean 2.17% vs. 3.02%, p = 0.008). In VHL-associated sHB, a higher MIB-1 index (≥ 2%) correlated with an increased risk of local tumor progression (AUC 0.74, 95% CI 0.49-0.98), whereas this was not observed in sporadic cases (AUC 0.56, 95% CI 0.23-0.88). Kaplan-Meier analysis showed that VHL patients with MIB-1 ≥ 2% had significantly shorter PFS (p = 0.05), while no significant association was found in sporadic tumors (p = 0.87). Our findings suggest that while VHL-associated sHB exhibit lower proliferative indices overall, elevated MIB-1 labeling indices might serve as a prognostic marker of shorter local PFS in this subgroup. In contrast, MIB-1 index appears to have limited prognostic relevance in sporadic sHB. These results highlight the importance of further molecular stratification and proliferation assessment in sHB to better inform clinical decision-making.</p>","PeriodicalId":6914,"journal":{"name":"Acta Neuropathologica Communications","volume":" ","pages":"18"},"PeriodicalIF":5.7,"publicationDate":"2025-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145740470","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}