Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Neurooncol ; 156(2): 295-306, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35001245

RESUMO

INTRODUCTION: The optimal treatment paradigm for brain metastasis that recurs locally after initial radiosurgery remains an area of active investigation. Here, we report outcomes for patients with BMRS treated with stereotactic laser ablation (SLA, also known as laser interstitial thermal therapy, LITT) followed by consolidation radiosurgery. METHODS: Clinical outcomes of 20 patients with 21 histologically confirmed BMRS treated with SLA followed by consolidation SRS and > 6 months follow-up were collected retrospectively across three participating institutions. RESULTS: Consolidation SRS (5 Gy × 5 or 6 Gy × 5) was carried out 16-73 days (median of 26 days) post-SLA in patients with BMRS. There were no new neurological deficits after SLA/cSRS. While 3/21 (14.3%) patients suffered temporary Karnofsky Performance Score (KPS) decline after SLA, no KPS decline was observed after cSRS. There were no 30-day mortalities or wound complications. Two patients required re-admission within 30 days of cSRS (severe headache that resolved with steroid therapy (n = 1) and new onset seizure (n = 1)). With a median follow-up of 228 days (range: 178-1367 days), the local control rate at 6 and 12 months (LC6, LC12) was 100%. All showed diminished FLAIR volume surrounding the SLA/cSRS treated BMRS at the six-month follow-up; none of the patients required steroid for symptoms attributable to these BMRS. These results compare favorably to the available literature for repeat SRS or SLA-only treatment of BMRS. CONCLUSIONS: This multi-institutional experience supports further investigations of SLA/cSRS as a treatment strategy for BMRS.


Assuntos
Neoplasias Encefálicas , Terapia a Laser , Recidiva Local de Neoplasia , Radiocirurgia , Técnicas de Ablação , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/radioterapia , Neoplasias Encefálicas/cirurgia , Terapia Combinada , Humanos , Terapia a Laser/métodos , Recidiva Local de Neoplasia/radioterapia , Recidiva Local de Neoplasia/cirurgia , Radiocirurgia/métodos , Estudos Retrospectivos , Técnicas Estereotáxicas , Resultado do Tratamento
2.
J Neurooncol ; 151(1): 3-12, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32542437

RESUMO

INTRODUCTION: The overall survival in patients with gliomas has not significantly increased in the modern era, despite advances such as immunotherapy. This is in part due to their notorious ability to suppress local and systemic immune responses, severely restricting treatment efficacy. METHODS: We have reviewed the preclinical and clinical evidence for immunosuppression seen throughout the disease process in gliomas. This review aims to discuss the various ways that brain tumors, and gliomas in particular, co-opt the body's immune system to evade detection and ensure tumor survival and proliferation. RESULTS: A multitude of mechanisms are discussed by which neoplastic cells evade detection and destruction by the immune system. These include tumor-induced T-cell and NK cell dysfunction, regulatory T-cell and myeloid-derived suppressor cell expansion, M2 phenotypic transformation in glioma-associated macrophages/microglia, upregulation of immunosuppressive glioma cell surface factors and cytokines, tumor microenvironment hypoxia, and iatrogenic sequelae of immunosuppressive treatments. CONCLUSIONS: Gliomas create a profoundly immunosuppressive environment, both locally within the tumor and systemically. Future research should aim to address these immunosuppressive mechanisms in the effort to generate treatment options with meaningful survival benefits for this patient population.


Assuntos
Neoplasias Encefálicas , Glioma , Humanos , Terapia de Imunossupressão , Macrófagos/imunologia , Microambiente Tumoral
3.
Int J Hyperthermia ; 37(2): 27-34, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32672126

RESUMO

The rise of immunotherapy (IT) in oncological treatment has greatly improved outcomes in a number of disease states. However, its use in tumors of the central nervous system (CNS) remains limited for multiple reasons related to the unique immunologic tumor microenvironment. As such, it is valuable to consider the intersection of IT with additional treatment methods that may improve access to the CNS and effectiveness of existing IT modalities. One such combination is the pairing of IT with localized hyperthermia (HT) generated through technologies such as laser interstitial thermal therapy (LITT). The wide-ranging immunomodulatory effects of localized and whole-body HT have been investigated for some time. Hyperthermia has demonstrated immunostimulatory effects at the level of tumor cells, immune cells, and the broader environment governing potential immune surveillance. A thorough understanding of these effects as well as the current and upcoming investigations of such in combination with IT is important in considering the future directions of neuro-oncology.


Assuntos
Neoplasias Encefálicas , Hipertermia Induzida , Imunoterapia , Terapia a Laser , Neoplasias Encefálicas/terapia , Humanos , Lasers , Microambiente Tumoral
4.
J Neurooncol ; 122(2): 293-301, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25579983

RESUMO

Myeloid derived suppressor cells (MDSCs) are bone marrow derived cells with immunosuppressive properties. We have shown previously that MDSCs numbers are elevated in the circulation of GBM patients and that they produce reversible T cell dysfunction. Here, we evaluated whether MDSCs infiltrate human GBM tissues, and whether a commonly used mouse model of GBM reproduces the biology of MDSCs that is observed in patients. We evaluated tumor specimens from patients with newly diagnosed GBM. We harvested and evaluated normal brain, tumors and hematopoietic tissues from control, vehicle and sunitinib-treated mice. In human GBM tumors, MDSCs represented 5.4 ± 1.8 % of total cells. The majority of MDSCs (CD33+HLADR-) were lineage negative (CD14-CD15-), followed by granulocytic (CD15+CD14-) and monocytic (CD15-CD14+) subtypes. In murine GBM tumors, MDSCs were 8.06 ± 0.78 % of total cells, of which more were monocytic (M-MDSC, CD11b+ Gr1-low) than granulocytic (G-MDSC, CD11b+ Gr1-high). Treatment with the tyrosine kinase inhibitor sunitinib decreased the infiltration of both granulocytic and monocytic MDSCs in murine GBM tumors. In the hematopoietic tissues, circulating G-MDSC blood levels were reduced after sunitinib treatment. In tumors, both CD3(+) and CD4(+) T cell counts increased following sunitinib treatment (p ≤ 0.001). Total T cell proliferation (p < 0.001) and interferon gamma production (p = 0.004) were increased in the spleens of sunitinib treated mice. Sunitinib-treated mice survived longer than vehicle-treated mice (p = 0.002). MDSCs are present in both human and mouse GBM tumors. Sunitinib may have an immunostimulatory effect, as its use is associated with a reduction in G-MDSCs and improvement in anti-tumor immune function.


Assuntos
Neoplasias Encefálicas/fisiopatologia , Encéfalo/fisiopatologia , Glioma/fisiopatologia , Linfócitos do Interstício Tumoral/fisiologia , Células Mieloides/fisiologia , Animais , Antineoplásicos/farmacologia , Medula Óssea/efeitos dos fármacos , Medula Óssea/fisiopatologia , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Linhagem Celular , Galinhas , Glioma/tratamento farmacológico , Glioma/patologia , Humanos , Indóis/farmacologia , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/patologia , Camundongos Transgênicos , Células Mieloides/efeitos dos fármacos , Células Mieloides/patologia , Transplante de Neoplasias , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Pirróis/farmacologia , Baço/efeitos dos fármacos , Baço/fisiopatologia , Sunitinibe , Análise de Sobrevida , Linfócitos T/efeitos dos fármacos , Linfócitos T/patologia , Linfócitos T/fisiologia , Resultado do Tratamento
5.
Clin Neuropathol ; 34(2): 76-82, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25601060

RESUMO

Meningioangiomatosis is a rare, benign, developmental, or hamartomatous lesion which may involve the leptomeninges and underlying brain parenchyma. Histologically, meningioangiomatosis is marked by a proliferation of blood vessels in the parenchyma, rimmed by collars of spindled meningothelial cells. There are anecdotal reports of an association of meningioangiomatosis with focal cortical dysplasia. We retrospectively analyzed the clinical, histopathologic, and treatment outcomes of 16 patients with a diagnosis of meningioangiomatosis, specifically investigating these cases for evidence of adjacent focal cortical dysplasia. Patients ranged in age from 1 to 34 years (median 18), 12 of whom had medically-intractable epilepsy as their presenting symptom. No patients in this study had a confirmed diagnosis of neurofibromatosis type II. Four patients (25%) were found to have fibrous meningiomas associated with the meningioangiomatosis. Ten of the 12 patients (83%) who had adequate tissue excised adjacent to the meningioangiomatosis demonstrated evidence of focal cortical dysplasia, with 6 of those (60%) classified as Palmini type IA, and 4 patients (40%) classified as Palmini type IIA. Seven of the patients (44%) had no post-operative seizures, and were off anti-epileptic drugs, while 2 patients relapsed, and required pharmacologic treatment for seizure control. This study therefore presents evidence to support inclusion of meningioangiomatosis as a focal cortical dysplasia-associated entity, as suggested by the ILAE classification (type IIIc). As focal cortical dysplasia is a developmental malformation, its association with meningioangiomatosis supports a developmental etiology of sporadic meningioangiomatosis.


Assuntos
Encefalopatias/complicações , Hamartoma/complicações , Malformações do Desenvolvimento Cortical/complicações , Adolescente , Adulto , Encefalopatias/patologia , Criança , Pré-Escolar , Feminino , Hamartoma/patologia , Humanos , Masculino , Malformações do Desenvolvimento Cortical/patologia , Estudos Retrospectivos , Adulto Jovem
7.
Clin Neuropathol ; 32(6): 515-21, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23863347

RESUMO

Calcifying pseudoneoplasms of the neuraxis (CPN), also known as fibroosseous lesions, are slow-growing lesions that can occur in a variety of regions of the central nervous system (CNS). These lesions are especially rare, with no more than 30 intracranial cases reported in the literature. Due to its non-malignant/non-metastatic course, one can expect an excellent prognosis with a successful complete resection. However, depending on the location of presentation within the CNS, CPN can present unique challenges in diagnosis and subsequently, the treatment course undertaken. We present a case of an intracranial, extra-axial CPN in a 49-year-old woman, the second to be reported in this location ever and first in 30 years, and detail the clinical, histopathologic and radiological features of a CPN along with a review of the literature.


Assuntos
Neoplasias Encefálicas/patologia , Calcinose/patologia , Corpo Caloso/patologia , Feminino , Humanos , Imageamento por Ressonância Magnética , Pessoa de Meia-Idade
8.
Neurosurgery ; 93(2): 427-435, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-36861990

RESUMO

BACKGROUND: Upfront laser interstitial thermal therapy (LITT) can be used as part of the treatment paradigm in difficult-to-access newly diagnosed glioblastoma multiforme (ndGBM) cases. The extent of ablation, though, is not routinely quantified; thus, its specific effect on patients' oncological outcomes is unclear. OBJECTIVE: To methodically measure the extent of ablation in the cohort of patients with ndGBM and its effect, and other treatment-related parameters, on patients' progression-free survival (PFS) and overall survival (OS). METHODS: A retrospective study was conducted on 56 isocitrate dehydrogenase 1/2 wild-type patients with ndGBM treated with upfront LITT between 2011 and 2021. Patient data including demographics, oncological course, and LITT-associated parameters were analyzed. RESULTS: Patient median age was 62.3 years (31-84), and the median follow-up duration was 11.4 months. As expected, the subgroup of patients receiving full chemoradiation was found to have the most beneficial PFS and OS (n = 34). Further analysis showed that 10 of them underwent near-total ablation and had a significantly improved PFS (10.3 months) and OS (22.7 months). Notably, 84% excess ablation was detected which was not related to a higher rate of neurological deficits. Tumor volume was also found to influence PFS and OS, but it was not possible to further corroborate this finding because of low numbers. CONCLUSION: This study presents data analysis of the largest series of ndGBM treated with upfront LITT. Near-total ablation was shown to significantly benefit patients' PFS and OS. Importantly, it was shown to be safe, even in cases of excess ablation and therefore could be considered when using this modality to treat ndGBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Terapia a Laser , Humanos , Pessoa de Meia-Idade , Glioblastoma/cirurgia , Estudos Retrospectivos , Resultado do Tratamento , Neoplasias Encefálicas/cirurgia , Lasers
9.
medRxiv ; 2023 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-36945620

RESUMO

Glioblastoma (GBM) treatment includes maximal safe resection of the core and MRI contrast-enhancing (CE) tumor. Complete resection of the infiltrative non-contrast-enhancing (NCE) tumor rim is rarely achieved. We established a safe, semi-automated workflow for spatially-registered sampling of MRI-defined GBM regions in 19 patients with downstream analysis and biobanking, enabling studies of NCE, wherefrom recurrence/progression typically occurs. Immunophenotyping revealed underrepresentation of myeloid cell subsets and CD8+ T cells in the NCE. While NCE T cells phenotypically and functionally resembled those in matching CE tumor, subsets of activated (CD69hi) effector memory CD8+ T cells were overrepresented. Contrarily, CD25hi Tregs and other subsets were underrepresented. Overall, our study demonstrated that MRI-guided, spatially-registered, intraoperative immunosampling is feasible as part of routine GBM surgery. Further elucidation of the shared and spatially distinct microenvironmental biology of GBM will enable development of therapeutic approaches targeting the NCE infiltrative tumor to decrease GBM recurrence.

10.
Cancer Discov ; 13(9): 2090-2105, 2023 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-37378557

RESUMO

Sex differences in glioblastoma (GBM) incidence and outcome are well recognized, and emerging evidence suggests that these extend to genetic/epigenetic and cellular differences, including immune responses. However, the mechanisms driving immunologic sex differences are not fully understood. Here, we demonstrate that T cells play a critical role in driving GBM sex differences. Male mice exhibited accelerated tumor growth, with decreased frequency and increased exhaustion of CD8+ T cells in the tumor. Furthermore, a higher frequency of progenitor exhausted T cells was found in males, with improved responsiveness to anti-PD-1 treatment. Moreover, increased T-cell exhaustion was observed in male GBM patients. Bone marrow chimera and adoptive transfer models indicated that T cell-mediated tumor control was predominantly regulated in a cell-intrinsic manner, partially mediated by the X chromosome inactivation escape gene Kdm6a. These findings demonstrate that sex-biased predetermined behavior of T cells is critical for inducing sex differences in GBM progression and immunotherapy response. SIGNIFICANCE: Immunotherapies in patients with GBM have been unsuccessful due to a variety of factors, including the highly immunosuppressive tumor microenvironment in GBM. This study demonstrates that sex-biased T-cell behaviors are predominantly intrinsically regulated, further suggesting sex-specific approaches can be leveraged to potentially improve the therapeutic efficacy of immunotherapy in GBM. See related commentary by Alspach, p. 1966. This article is featured in Selected Articles from This Issue, p. 1949.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Masculino , Feminino , Camundongos , Animais , Glioblastoma/genética , Exaustão das Células T , Linfócitos T CD8-Positivos , Imunoterapia , Imunidade , Neoplasias Encefálicas/patologia , Microambiente Tumoral
11.
bioRxiv ; 2023 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-38014234

RESUMO

The glioblastoma microenvironment is enriched in immunosuppressive factors that potently interfere with the function of cytotoxic T lymphocytes. Cancer cells can directly impact the immune system, but the mechanisms driving these interactions are not completely clear. Here we demonstrate that the polyamine metabolite spermidine is elevated in the glioblastoma tumor microenvironment. Exogenous administration of spermidine drives tumor aggressiveness in an immune-dependent manner in pre-clinical mouse models via reduction of CD8+ T cell frequency and phenotype. Knockdown of ornithine decarboxylase, the rate-limiting enzyme in spermidine synthesis, did not impact cancer cell growth in vitro but did result in extended survival. Furthermore, glioblastoma patients with a more favorable outcome had a significant reduction in spermidine compared to patients with a poor prognosis. Our results demonstrate that spermidine functions as a cancer cell-derived metabolite that drives tumor progression by reducing CD8+T cell number and function.

12.
JCI Insight ; 8(13)2023 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-37252795

RESUMO

Intratumoral heterogeneity is a defining hallmark of glioblastoma, driving drug resistance and ultimately recurrence. Many somatic drivers of microenvironmental change have been shown to affect this heterogeneity and, ultimately, the treatment response. However, little is known about how germline mutations affect the tumoral microenvironment. Here, we find that the single-nucleotide polymorphism (SNP) rs755622 in the promoter of the cytokine macrophage migration inhibitory factor (MIF) is associated with increased leukocyte infiltration in glioblastoma. Furthermore, we identified an association between rs755622 and lactotransferrin expression, which could also be used as a biomarker for immune-infiltrated tumors. These findings demonstrate that a germline SNP in the promoter region of MIF may affect the immune microenvironment and further reveal a link between lactotransferrin and immune activation.


Assuntos
Glioblastoma , Fatores Inibidores da Migração de Macrófagos , Humanos , Lactoferrina/genética , Fatores Inibidores da Migração de Macrófagos/genética , Polimorfismo de Nucleotídeo Único , Glioblastoma/genética , Regiões Promotoras Genéticas , Microambiente Tumoral/genética , Oxirredutases Intramoleculares/genética
13.
J Clin Invest ; 132(8)2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35426370

RESUMO

Given its aggressive natural history and immunosuppressive nature, glioblastoma (GBM) remains difficult to treat. Tumor Treating Fields (TTFields) are a promising treatment for GBM patients, yet the entirety of their antitumor action has not been fully elucidated. In a recent issue of the JCI, Chen et al. explored the effect of TTFields in reinvigorating immune responses. By elegant step-by-step approaches, the authors demonstrated that TTFields promote the production of immune-stimulating proinflammatory and interferon type 1 cytokines in tumor cells in a cGAS/STING- and AIM2 inflammasome-dependent mechanism, thereby activating the immune system. The findings show that TTFields not only directly inhibit tumor cell growth, as previously reported, but enhance antitumor immunity, suggesting TTFields can be used as an immune-modulating approach in GBM.


Assuntos
Glioblastoma , Ciclo Celular , Proliferação de Células , Glioblastoma/patologia , Glioblastoma/terapia , Humanos
14.
Neurosurgery ; 90(6): 684-690, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35311745

RESUMO

BACKGROUND: Radiation necrosis (RN) after stereotactic radiosurgery (SRS) for brain metastases (BM) can result in significant morbidity, compounded by the effects of extended steroid therapy. Laser interstitial thermal therapy (LITT) is a minimally invasive procedure that can offer definitive treatment for RN while potentially obviating the need for prolonged steroid use. OBJECTIVE: To compare LITT vs medical management (MM) in the treatment of RN. METHODS: A multicenter, retrospective study was performed of SRS-treated patients with BM who developed biopsy-proven RN and were treated with LITT or MM. Clinical outcome data were compared by treatment modality. RESULTS: Seventy-two patients met criteria with a median follow-up of 10.0 months (4.2-25.1), and 57 patients (79%) underwent LITT. Four MM (27%) and 3 LITT patients (5%) demonstrated radiographic progression (P = .031) at a median of 5.3 and 4.0 months (P = .40). There was no significant difference in overall survival (LITT median of 15.2 vs 11.6 months, P = .60) or freedom from local progression (13.6 vs 7.06 months, P = .40). Patients stopped steroid therapy earlier in the LITT cohort at a median of 37 days compared with 245 days (P < .001). When controlled for follow-up duration, patients treated with LITT were 3 times more likely to be weaned off steroids before the study end point (P = .003). CONCLUSION: These data suggest that LITT for treatment of biopsy-proven RN after SRS for BM significantly decreases time to steroid independence. Prospective trials should be designed to further validate the utility of LITT for RN and its impact on steroid-induced morbidity.


Assuntos
Neoplasias Encefálicas , Terapia a Laser , Lesões por Radiação , Radiocirurgia , Biópsia , Neoplasias Encefálicas/patologia , Humanos , Terapia a Laser/efeitos adversos , Terapia a Laser/métodos , Lasers , Necrose/etiologia , Necrose/cirurgia , Recidiva Local de Neoplasia/cirurgia , Estudos Prospectivos , Lesões por Radiação/terapia , Radiocirurgia/efeitos adversos , Radiocirurgia/métodos , Estudos Retrospectivos , Esteroides/uso terapêutico , Resultado do Tratamento
15.
Cancer Res ; 82(22): 4274-4287, 2022 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-36126163

RESUMO

In multiple types of cancer, an increased frequency in myeloid-derived suppressor cells (MDSC) is associated with worse outcomes and poor therapeutic response. In the glioblastoma (GBM) microenvironment, monocytic (m) MDSCs represent the predominant subset. However, the molecular basis of mMDSC enrichment in the tumor microenvironment compared with granulocytic (g) MDSCs has yet to be determined. Here we performed the first broad epigenetic profiling of MDSC subsets to define underlying cell-intrinsic differences in behavior and found that enhanced gene accessibility of cell adhesion programs in mMDSCs is linked to their tumor-accelerating ability in GBM models upon adoptive transfer. Mouse and human mMDSCs expressed higher levels of integrin ß1 and dipeptidyl peptidase-4 (DPP-4) compared with gMDSCs as part of an enhanced cell adhesion signature. Integrin ß1 blockade abrogated the tumor-promoting phenotype of mMDSCs and altered the immune profile in the tumor microenvironment, whereas treatment with a DPP-4 inhibitor extended survival in preclinical GBM models. Targeting DPP-4 in mMDSCs reduced pERK signaling and their migration towards tumor cells. These findings uncover a fundamental difference in the molecular basis of MDSC subsets and suggest that integrin ß1 and DPP-4 represent putative immunotherapy targets to attenuate myeloid cell-driven immune suppression in GBM. SIGNIFICANCE: Epigenetic profiling uncovers cell adhesion programming as a regulator of the tumor-promoting functions of monocytic myeloid-derived suppressor cells in glioblastoma, identifying therapeutic targets that modulate the immune response and suppress tumor growth.


Assuntos
Adesão Celular , Glioblastoma , Células Supressoras Mieloides , Animais , Humanos , Camundongos , Glioblastoma/metabolismo , Glioblastoma/patologia , Integrina beta1/metabolismo , Células Supressoras Mieloides/patologia , Microambiente Tumoral
16.
Neurooncol Adv ; 4(1): vdac086, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35795470

RESUMO

Background: Improved survival for patients with brain metastases has been accompanied by a rise in tumor recurrence after stereotactic radiotherapy (SRT). Laser interstitial thermal therapy (LITT) has emerged as an effective treatment for SRT failures as an alternative to open resection or repeat SRT. We aimed to evaluate the efficacy of LITT followed by SRT (LITT+SRT) in recurrent brain metastases. Methods: A multicenter, retrospective study was performed of patients who underwent treatment for biopsy-proven brain metastasis recurrence after SRT at an academic medical center. Patients were stratified by "planned LITT+SRT" versus "LITT alone" versus "repeat SRT alone." Index lesion progression was determined by modified Response Assessment in Neuro-Oncology Brain Metastases (RANO-BM) criteria. Results: Fifty-five patients met inclusion criteria, with a median follow-up of 7.3 months (range: 1.0-30.5), age of 60 years (range: 37-86), Karnofsky Performance Status (KPS) of 80 (range: 60-100), and pre-LITT/biopsy contrast-enhancing volume of 5.7 cc (range: 0.7-19.4). Thirty-eight percent of patients underwent LITT+SRT, 45% LITT alone, and 16% SRT alone. Median time to index lesion progression (29.8, 7.5, and 3.7 months [P = .022]) was significantly improved with LITT+SRT. When controlling for age in a multivariate analysis, patients treated with LITT+SRT remained significantly less likely to have index lesion progression (P = .004). Conclusions: These data suggest that LITT+SRT is superior to LITT or repeat SRT alone for treatment of biopsy-proven brain metastasis recurrence after SRT failure. Prospective trials are warranted to validate the efficacy of using combination LITT+SRT for treatment of recurrent brain metastases.

17.
Neurosurg Clin N Am ; 32(1): 105-115, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33223019

RESUMO

The previous decade has seen an expansion in the use of laser interstitial thermal therapy (LITT) for a variety of pathologies. LITT has been used to treat both newly diagnosed and recurrent glioblastoma (GBM), especially in deep-seated, difficult-to-access lesions where open resection is otherwise infeasible or in patients who would not tolerate craniotomy. This review aims to describe the current state of the technology and operative technique, as well as summarize the outcomes data and future research regarding LITT as a treatment of GBM.


Assuntos
Neoplasias Encefálicas/cirurgia , Glioblastoma/cirurgia , Terapia a Laser/métodos , Humanos , Procedimentos Cirúrgicos Minimamente Invasivos/métodos , Técnicas Estereotáxicas , Resultado do Tratamento
18.
Neurooncol Adv ; 3(Suppl 5): v16-v25, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34859229

RESUMO

Laser interstitial thermal therapy (LITT) is a minimally invasive treatment for intracranial lesions entailing thermal ablation via a stereotactically placed laser probe. In metastatic disease, it has shown the most promise in the treatment of radiographically progressive lesions after initial stereotactic radiosurgery, whether due to recurrent metastatic disease or radiation necrosis. LITT has been demonstrated to provide clinical benefit in both cases, as discussed in the review below. With its minimal surgical footprint and short recovery period, LITT is further advantaged for patients who are otherwise high-risk surgical candidates or with lesions in difficult to access locations. Exploration of the current data on its use in metastatic disease will allow for a better understanding of the indications, benefits, and future directions of LITT for these patients.

19.
J Neurosurg Spine ; 34(6): 864-870, 2021 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-33823491

RESUMO

OBJECTIVE: In a healthcare landscape in which costs increasingly matter, the authors sought to distinguish among the clinical and nonclinical drivers of patient length of stay (LOS) in the hospital following elective lumbar laminectomy-a common spinal surgery that may be reimbursed using bundled payments-and to understand their relationships with patient outcomes and costs. METHODS: Patients ≥ 18 years of age undergoing laminectomy surgery for degenerative lumbar spinal stenosis within the Cleveland Clinic health system between March 1, 2016, and February 1, 2019, were included in this analysis. Generalized linear modeling was used to assess the relationships between the day of surgery, patient discharge disposition, and hospital LOS, while adjusting for underlying patient health risks and other nonclinical factors, including the hospital surgery site and health insurance. RESULTS: A total of 1359 eligible patients were included in the authors' analysis. The mean LOS ranged between 2.01 and 2.47 days for Monday and Friday cases, respectively. The LOS was also notably longer for patients who were ultimately discharged to a skilled nursing facility (SNF) or rehabilitation center. A prolonged LOS occurring later in the week was not associated with greater underlying health risks, yet it nevertheless resulted in greater costs of care: the average total surgical costs for lumbar laminectomy were 20% greater for Friday cases than for Monday cases, and 24% greater for late-week cases than for early-week cases ultimately transferred to SNFs or rehabilitation centers. A Poisson generalized linear model fit the data best and showed that the comorbidity burden, surgery at a tertiary care center versus a community hospital, and the incidence of any postoperative complication were associated with significantly longer hospital stays. Discharge to home healthcare, SNFs, or rehabilitation centers, and late-week surgery were significant nonclinical predictors of LOS prolongation, even after adjusting for underlying patient health risks and insurance, with LOSs that were, for instance, 1.55 and 1.61 times longer for patients undergoing their procedure on Thursday and Friday compared to Monday, respectively. CONCLUSIONS: Late-week surgeries are associated with a prolonged LOS, particularly when discharge is to an SNF or rehabilitation center. These findings point to opportunities to lower costs and improve outcomes associated with elective surgical care. Interventions to optimize surgical scheduling and perioperative care coordination could help reduce prolonged LOSs, lower costs, and, ultimately, give service line management personnel greater flexibility over how to use existing resources as they remain ahead of healthcare reforms.

20.
Clin Cancer Res ; 27(7): 2038-2049, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33542075

RESUMO

PURPOSE: Glioblastoma (GBM) immunotherapy clinical trials are generally initiated after standard-of-care treatment-including surgical resection, perioperative high-dose steroid therapy, chemotherapy, and radiation treatment-has either begun or failed. However, the impact of these interventions on the antitumoral immune response is not well studied. While discoveries regarding the impact of chemotherapy and radiation on immune response have been made and translated into clinical trial design, the impact of surgical resection and steroids on the antitumor immune response has yet to be determined. EXPERIMENTAL DESIGN: We developed a murine model integrating tumor resection and steroid treatment and used flow cytometry to analyze systemic and local immune changes. These mouse model findings were validated in a cohort of 95 patients with primary GBM. RESULTS: Using our murine resection model, we observed a systemic reduction in lymphocytes corresponding to increased tumor volume and decreased circulating lymphocytes that was masked by dexamethasone treatment. The reduction in circulating T cells was due to reduced CCR7 expression, resulting in T-cell sequestration in lymphoid organs and the bone marrow. We confirmed these findings in a cohort of patients with primary GBM and found that prior to steroid treatment, circulating lymphocytes inversely correlated with tumor volume. Finally, we demonstrated that peripheral lymphocyte content varies with progression-free survival and overall survival, independent of tumor volume, steroid use, or molecular profiles. CONCLUSIONS: These data reveal that prior to intervention, increased tumor volume corresponds with reduced systemic immune function and that peripheral lymphocyte counts are prognostic when steroid treatment is taken into account.


Assuntos
Neoplasias Encefálicas/imunologia , Glioblastoma/imunologia , Idoso , Animais , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Dexametasona/uso terapêutico , Modelos Animais de Doenças , Feminino , Glioblastoma/mortalidade , Glioblastoma/patologia , Glioblastoma/terapia , Humanos , Tolerância Imunológica , Imunofenotipagem , Contagem de Linfócitos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Carga Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA