Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
2.
Technol Cancer Res Treat ; 21: 15330338221124658, 2022.
Article in English | MEDLINE | ID: mdl-36172744

ABSTRACT

Introduction: Although there have been significant advances in research and treatments over the past decades, cancer remains a leading cause of morbidity and mortality, mostly due to resistance to standard therapies. Pulsed electromagnetic field (PEMF), a newly emerged therapeutic strategy, has been highly regarded as less invasive and almost safe to patients, is now a clinically accepted form to treat diseases including cancer. Breast and lung cancer are the most prevalent forms of human cancers, yet reported investigations on exploring regimes including PEMF are limited. Methods: Intended to examine the anti-tumor effects of a clinically accepted osteogenic PEMF and the possibility of including PEMF in breast and lung cancer treatments, we studied the effects of 2 PEMF signals (PMF1 and PMF2) on breast and lung cancer cell growth and proliferation, as well as the possible underline mechanisms in vitro and in vivo. Results: We found that both signals caused modest but significant growth inhibition (∼5%) in MCF-7 and A549 cancer cells. Interestingly, mice xenograft tumors with A549 cells treated by PEMF were smaller in sizes than controls. However, for mice with MCF-7 tumor implants, treatment with PMF1 resulted in a slight increase (2.8%) in mean tumor size, while PMF2 treated tumors showed a 9% reduction in average size. Furthermore, PEMF increased caspase 3/7 expression levels and percentage of annexin stained cells, indicating the induction of apoptosis. It also increased G0 by 8.5%, caused changes in the expression of genes associated with cell growth suppression, DNA damage, cell cycle arrest, and apoptosis. When cancer cells or xenograft tumors treated with combined PEMF and chemotherapy drugs, PEMF showed growth inhibition effect independent of cisplatin in A549 cells, but with added effect by pemetrexed for the inhibition of MCF-7 growth. Conclusion: Together, our data suggested that clinically used osteogenic PEMF signals moderately suppressed cancer cell growth and proliferation both in vitro and in vivo.


Subject(s)
Breast Neoplasms , Electromagnetic Fields , Lung Neoplasms , A549 Cells , Animals , Annexins , Breast Neoplasms/therapy , Caspase 3 , Cisplatin , Humans , Lung Neoplasms/therapy , MCF-7 Cells , Mice , Pemetrexed
3.
Sci Rep ; 11(1): 16866, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34654856

ABSTRACT

Prognosis of patients with HER2+ breast-to-brain-metastasis (BBM) is dismal even after current standard-of-care treatments, including surgical resection, whole-brain radiation, and systemic chemotherapy. Radiation and systemic chemotherapies can also induce cytotoxicity, leading to significant side effects. Studies indicate that donor-derived platelets can serve as immune-compatible drug carriers that interact with and deliver drugs to cancer cells with fewer side effects, making them a promising therapeutic option with enhanced antitumor activity. Moreover, human induced pluripotent stem cells (hiPSCs) provide a potentially renewable source of clinical-grade transfusable platelets that can be drug-loaded to complement the supply of donor-derived platelets. Here, we describe methods for ex vivo generation of megakaryocytes (MKs) and functional platelets from hiPSCs (hiPSC-platelets) in a scalable fashion. We then loaded hiPSC-platelets with lapatinib and infused them into BBM tumor-bearing NOD/SCID mouse models. Such treatment significantly increased intracellular lapatinib accumulation in BBMs in vivo, potentially via tumor cell-induced activation/aggregation. Lapatinib-loaded hiPSC-platelets exhibited normal morphology and function and released lapatinib pH-dependently. Importantly, lapatinib delivery to BBM cells via hiPSC-platelets inhibited tumor growth and prolonged survival of tumor-bearing mice. Overall, use of lapatinib-loaded hiPSC-platelets effectively reduced adverse effects of free lapatinib and enhanced its therapeutic efficacy, suggesting that they represent a novel means to deliver chemotherapeutic drugs as treatment for BBM.


Subject(s)
Brain Neoplasms/drug therapy , Breast Neoplasms/drug therapy , Induced Pluripotent Stem Cells/drug effects , Lapatinib/pharmacology , Neoplasm Metastasis/pathology , Receptor, ErbB-2/drug effects , Animals , Antineoplastic Agents/pharmacology , Brain Neoplasms/secondary , Breast Neoplasms/pathology , Drug Carriers/pharmacology , Humans , Induced Pluripotent Stem Cells/cytology , Mice, Inbred NOD , Mice, SCID , Quinazolines/pharmacology , Receptor, ErbB-2/metabolism
4.
Cancer Res ; 81(12): 3200-3214, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33941612

ABSTRACT

HER2+ breast leptomeningeal carcinomatosis (HER2+ LC) occurs when tumor cells spread to cerebrospinal fluid-containing leptomeninges surrounding the brain and spinal cord, a complication with a dire prognosis. HER2+ LC remains incurable, with few treatment options. Currently, much effort is devoted toward development of therapies that target mutations. However, targeting epigenetic or transcriptional states of HER2+ LC tumors might efficiently target HER2+ LC growth via inhibition of oncogenic signaling; this approach remains promising but is less explored. To test this possibility, we established primary HER2+ LC (Lepto) cell lines from nodular HER2+ LC tissues. These lines are phenotypically CD326+CD49f-, confirming that they are derived from HER2+ LC tumors, and express surface CD44+CD24-, a cancer stem cell (CSC) phenotype. Like CSCs, Lepto lines showed greater drug resistance and more aggressive behavior compared with other HER2+ breast cancer lines in vitro and in vivo. Interestingly, the three Lepto lines overexpressed Jumonji domain-containing histone lysine demethylases KDM4A/4C. Treatment with JIB04, a selective inhibitor of Jumonji demethylases, or genetic loss of function of KDM4A/4C induced apoptosis and cell-cycle arrest and reduced Lepto cell viability, tumorsphere formation, regrowth, and invasion in vitro. JIB04 treatment of patient-derived xenograft mouse models in vivo reduced HER2+ LC tumor growth and prolonged animal survival. Mechanistically, KDM4A/4C inhibition downregulated GMCSF expression and prevented GMCSF-dependent Lepto cell proliferation. Collectively, these results establish KDM4A/4C as a viable therapeutic target in HER2+ LC and spotlight the benefits of targeting the tumorigenic transcriptional network. SIGNIFICANCE: HER2+ LC tumors overexpress KDM4A/4C and are sensitive to the Jumonji demethylase inhibitor JIB04, which reduces the viability of primary HER2+ LC cells and increases survival in mouse models.


Subject(s)
Aminopyridines/pharmacology , Breast Neoplasms/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hydrazones/pharmacology , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Meningeal Carcinomatosis/drug therapy , Receptor, ErbB-2/metabolism , Animals , Apoptosis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Female , Humans , Meningeal Carcinomatosis/metabolism , Meningeal Carcinomatosis/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Clin Exp Metastasis ; 37(3): 401-412, 2020 06.
Article in English | MEDLINE | ID: mdl-32279122

ABSTRACT

The brain is often reported as the first site of recurrence among breast cancer patients overexpressing human epidermal growth factor receptor 2 (HER2). Although most HER2+tumors metastasize to the subcortical region of the brain, a subset develops in the cortical region. We hypothesize that factors in cerebrospinal fluid (CSF) play a critical role in the adaptation, proliferation, and establishment of cortical metastases. We established novel cell lines using patient biopsies to model breast cancer cortical and subcortical metastases. We assessed the localization and growth of these cells in vivo and proliferation and apoptosis in vitro under various conditions. Proteomic analysis of human CSF identified astrocyte-derived factors that support the proliferation of cortical metastases, and we used neutralizing antibodies to test the effects of inhibiting these factors both in vivo and in vitro. The cortical breast cancer brain metastatic cells exhibited greater proliferation than subcortical breast cancer brain metastatic cells in CSF containing several growth factors that nourish both the CNS and tumor cells. Specifically, the astrocytic paracrine factors IGFBP2 and CHI3LI promoted the proliferation of cortical metastatic cells and the formation of metastatic lesions. Disruption of these factors suppressed astrocyte-tumor cell interactions in vitro and the growth of cortical tumors in vivo. Our findings suggest that inhibition of IGFBP2 and CHI3LI signaling, in addition to existing treatment modalities, may be an effective therapeutic strategy targeting breast cancer cortical metastasis.


Subject(s)
Astrocytes/pathology , Brain Neoplasms/secondary , Breast Neoplasms/pathology , Cerebrospinal Fluid/cytology , Chitinase-3-Like Protein 1/metabolism , Insulin-Like Growth Factor Binding Protein 2/metabolism , Animals , Apoptosis/drug effects , Brain Neoplasms/cerebrospinal fluid , Brain Neoplasms/drug therapy , Breast Neoplasms/cerebrospinal fluid , Cell Proliferation/drug effects , Cerebral Cortex/pathology , Chitinase-3-Like Protein 1/antagonists & inhibitors , Coculture Techniques , Female , Humans , Insulin-Like Growth Factor Binding Protein 2/antagonists & inhibitors , Mice , Paracrine Communication , Primary Cell Culture , Proteomics , Receptor, ErbB-2/metabolism , Signal Transduction/drug effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Sci Rep ; 9(1): 197, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30655550

ABSTRACT

Chromobox 6 (CBX6) is a subunit of Polycomb Repressive Complex 1 (PRC1) that mediates epigenetic gene repression and acts as an oncogene or tumor suppressor in a cancer type-dependent manner. The specific function of CBX6 in breast cancer is currently undefined. In this study, a comprehensive analysis of The Cancer Genome Atlas (TCGA) dataset led to the identification of CBX6 as a consistently downregulated gene in breast cancer. We provided evidence showing enhancer of zeste homolog 2 (EZH2) negatively regulated CBX6 expression in a Polycomb Repressive Complex 2 (PRC2)-dependent manner. Exogenous overexpression of CBX6 inhibited cell proliferation and colony formation, and induced cell cycle arrest along with suppression of migration and invasion of breast cancer cells in vitro. Microarray analyses revealed that CBX6 governs a complex gene expression program. Moreover, CBX6 induced significant downregulation of bone marrow stromal cell antigen-2 (BST2), a potential therapeutic target, via interactions with its promoter region. Our collective findings support a tumor suppressor role of CBX6 in breast cancer.


Subject(s)
Breast Neoplasms/pathology , Enhancer of Zeste Homolog 2 Protein/physiology , Genes, Tumor Suppressor , Polycomb-Group Proteins/physiology , Antigens, CD/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Down-Regulation , GPI-Linked Proteins/metabolism , Gene Expression Regulation, Neoplastic , Humans , Polycomb-Group Proteins/genetics
8.
Int J Mol Sci ; 19(2)2018 Jan 30.
Article in English | MEDLINE | ID: mdl-29385725

ABSTRACT

Cancers that exhibit the Warburg effect may elevate expression of glyoxylase 1 (GLO1) to detoxify the toxic glycolytic byproduct methylglyoxal (MG) and inhibit the formation of pro-apoptotic advanced glycation endproducts (AGEs). Inhibition of GLO1 in cancers that up-regulate glycolysis has been proposed as a therapeutic targeting strategy, but this approach has not been evaluated for glioblastoma multiforme (GBM), the most aggressive and difficult to treat malignancy of the brain. Elevated GLO1 expression in GBM was established in patient tumors and cell lines using bioinformatics tools and biochemical approaches. GLO1 inhibition in GBM cell lines and in an orthotopic xenograft GBM mouse model was examined using both small molecule and short hairpin RNA (shRNA) approaches. Inhibition of GLO1 with S-(p-bromobenzyl) glutathione dicyclopentyl ester (p-BrBzGSH(Cp)2) increased levels of the DNA-AGE N²-1-(carboxyethyl)-2'-deoxyguanosine (CEdG), a surrogate biomarker for nuclear MG exposure; substantially elevated expression of the immunoglobulin-like receptor for AGEs (RAGE); and induced apoptosis in GBM cell lines. Targeting GLO1 with shRNA similarly increased CEdG levels and RAGE expression, and was cytotoxic to glioma cells. Mice bearing orthotopic GBM xenografts treated systemically with p-BrBzGSH(Cp)2 exhibited tumor regression without significant off-target effects suggesting that GLO1 inhibition may have value in the therapeutic management of these drug-resistant tumors.


Subject(s)
Brain Neoplasms , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma , Lactoylglutathione Lyase , Neoplasm Proteins , Receptor for Advanced Glycation End Products/biosynthesis , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Heterografts , Humans , Lactoylglutathione Lyase/antagonists & inhibitors , Lactoylglutathione Lyase/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Xenograft Model Antitumor Assays
9.
Clin Exp Metastasis ; 34(2): 185-196, 2017 02.
Article in English | MEDLINE | ID: mdl-28210910

ABSTRACT

Breast cancer metastasis to the brain develops after a clinical latency of years to even decades, suggesting that colonization of the brain is the most challenging step of the metastatic cascade. However, the underlying mechanisms used by breast cancer cells to successfully colonize the brain's microenvironment remain elusive. Reelin is an archetypal extracellular glycoprotein that regulates migration, proliferation, and lamination of neurons. It is epigenetically silenced in various cancers, and its expression in multiple myelomas is linked to poor patient survival. We found that Reelin expression was low in primary breast cancer tissue. However, its expression was significantly higher in Her2+ breast cancers metastasizing to the brain. In particular, Reelin was highly expressed in the tumor periphery adjacent to surrounding astrocytes. This augmented Reelin expression was seen in Her2+ metastases, but not in triple negative (TN) primary tumors or in TN breast to brain metastasis cells co-cultured with astrocytes. Furthermore, the elevated expression was sustained in Her2+ cells grown in the presence of the DNA methyltransferase inhibitor 5-azacytidine, indicating epigenetic regulation of Reelin expression. The relative growth and rate of spheroids formation derived from Her2+ primary and BBM cells co-cultured with astrocytes were higher than those of TN primary and BBM cells, and knockdown of both Reelin and Her2 suppressed the astrocyte-induced growth and spheroid forming ability of Her2+ cells. Collectively, our results indicate that within the neural niche, astrocytes epigenetically regulate Reelin expression and its interaction with Her2 leading to increased proliferation and survival fitness.


Subject(s)
Astrocytes/metabolism , Brain Neoplasms/secondary , Breast Neoplasms/pathology , Cell Adhesion Molecules, Neuronal/physiology , Extracellular Matrix Proteins/physiology , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/physiology , Nerve Tissue Proteins/physiology , Receptor, ErbB-2/physiology , Serine Endopeptidases/physiology , Azacitidine/pharmacology , Brain Neoplasms/blood supply , Brain Neoplasms/metabolism , Breast Neoplasms/genetics , Cell Adhesion Molecules, Neuronal/biosynthesis , Cell Adhesion Molecules, Neuronal/genetics , Cell Division , Coculture Techniques , Culture Media, Serum-Free , DNA Methylation/drug effects , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , RNA Interference , RNA, Small Interfering/genetics , Reelin Protein , Serine Endopeptidases/biosynthesis , Serine Endopeptidases/genetics , Spheroids, Cellular/drug effects , Transcriptional Activation , Triple Negative Breast Neoplasms/secondary , Tumor Cells, Cultured
10.
J Neurosurg ; 124(5): 1310-8, 2016 May.
Article in English | MEDLINE | ID: mdl-26544779

ABSTRACT

OBJECT Despite recent advances, metastatic melanoma remains a terminal disease, in which life-threatening brain metastasis occurs in approximately half of patients. Sorafenib is a multikinase inhibitor that induces apoptosis of melanoma cells in vitro. However, systemic administration has been ineffective because adequate tissue concentrations cannot be achieved. This study investigated if convection-enhanced delivery (CED) of sorafenib would enhance tumor control and survival via inhibition of the signal transducer and activator of transcription 3 (Stat3) pathway in a murine model of metastatic brain melanoma. METHODS Melanoma cells treated with sorafenib in vitro were examined for signaling and survival changes. The effect of sorafenib given by CED was assessed by bioluminescent imaging and animal survival. RESULTS The results showed that sorafenib induced cell death in the 4 established melanoma cell lines and in 1 primary cultured melanoma cell line. Sorafenib inhibited Stat3 phosphorylation in HTB65, WYC1, and B16 cells. Accordingly, sorafenib treatment also decreased expression of Mcl-1 mRNA in melanoma cell lines. Because sorafenib targets multiple pathways, the present study demonstrated the contribution of the Stat3 pathway by showing that mouse embryonic fibroblast (MEF) Stat3 +/+ cells were significantly more sensitive to sorafenib than MEF Stat3 -/- cells. In the murine model of melanoma brain metastasis used in this study, CED of sorafenib increased survival by 150% in the treatment group compared with animals receiving the vehicle control (p < 0.01). CED of sorafenib also significantly abrogated tumor growth. CONCLUSIONS The data from this study indicate that local delivery of sorafenib effectively controls brain melanoma. These findings validate further investigation of the use of CED to distribute molecularly targeted agents.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/secondary , Convection , Melanoma, Experimental/drug therapy , Melanoma, Experimental/secondary , Niacinamide/analogs & derivatives , Phenylurea Compounds/administration & dosage , Phenylurea Compounds/pharmacology , STAT3 Transcription Factor/genetics , Transcriptional Activation/drug effects , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Heterografts , Humans , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Niacinamide/administration & dosage , Niacinamide/pharmacology , Sorafenib
11.
World Neurosurg ; 85: 42-8, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26260940

ABSTRACT

OBJECTIVE: Bony metastasis predominantly affects the spinal column and has been commonly associated in patients with breast cancer. There are two types of lesions that can occur with spine cancer-osteolytic or osteoblastic. Some patients may have mixed lesions, which include lytic and blastic in one vertebra or lytic and blastic in different vertebrae. Previous studies have shown that patients with breast cancer have an increased likelihood for development of lytic spinal metastases. METHODS: A retrospective chart review was conducted to more closely examine the association between hormone receptor status and spinal lesion type. A total of 195 patients were initially identified through the City of Hope Cancer Registry. Of the 195, only 153 patients had hormone receptor marker status available. Associations between spinal lesion and hormone receptor status were evaluated using χ(2) tests with alpha = 0.05 significance level. In a secondary analysis, the Oncomine Platform was used, which integrated The Cancer Genome Atlas (TCGA) datasets, to identify osteogenic genes that may be relevant to invasive breast cancers. RESULTS: Contrary to previous studies, our findings revealed progesterone receptor positive (PR+) patients were significantly more likely to present with blastic than lytic or mixed lesions. Furthermore, using TCGA analysis, COL1A1 and COL1A2 were found to be up-regulated, which could provide a molecular explanation for the development of blastic metastases. CONCLUSIONS: By integrating clinical and bioinformatic techniques, this study provides a novel discovery of the relationship between blastic and PR + breast cancers, which may have important implications for diagnostic strategies concerning vertebral metastases.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Collagen Type I/genetics , Receptor, ErbB-2/genetics , Receptors, Estrogen/genetics , Receptors, Progesterone/genetics , Spinal Neoplasms/genetics , Spinal Neoplasms/secondary , Adult , Aged , Aged, 80 and over , Breast/pathology , Collagen Type I, alpha 1 Chain , Female , Humans , Middle Aged , Osteoblasts/pathology , Osteoclasts/pathology , Retrospective Studies , Spinal Neoplasms/pathology , Spine/pathology , Up-Regulation/genetics
14.
Clin Exp Metastasis ; 30(6): 753-68, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23456474

ABSTRACT

Brain colonization by metastatic tumor cells offers a unique opportunity to investigate microenvironmental influences on the neoplastic process. The bi-directional interplay of breast cancer cells (mesodermal origin) and brain cells (neuroectodermal origin) is poorly understood and rarely investigated. In our patients undergoing neurosurgical resection of breast-to-brain metastases, specimens from the tumor/brain interface exhibited increased active gliosis as previously described. In addition, our histological characterization revealed infiltration of neural progenitor cells (NPCs) both outside and inside the tumor margin, leading us to investigate the cellular and molecular interactions between NPCs and metastases. Since signaling by the TGF-ß superfamily is involved in both developmental neurobiology and breast cancer pathogenesis, we examined the role of these proteins in the context of brain metastases. The brain-metastatic breast cancer cell line MDA-MB-231Br (231Br) expressed BMP-2 at significantly higher levels compared to its matched primary breast cancer cell line MDA-MB-231 (231). Co-culturing was used to examine bi-directional cellular effects and the relevance of BMP-2 overexpression. When co-cultured with NPCs, 231 (primary) tumor cells failed to proliferate over 15 days. However, 231Br (brain metastatic) tumor cells co-cultured with NPCs escaped growth inhibition after day 5 and proliferated, occurring in parallel with NPC differentiation into astrocytes. Using shRNA and gene knock-in, we then demonstrated BMP-2 secreted by 231Br cells mediated NPC differentiation into astrocytes and concomitant tumor cell proliferation in vitro. In xenografts, overexpression of BMP-2 in primary breast cancer cells significantly enhanced their ability to engraft and colonize the brain, thereby creating a metastatic phenotype. Conversely, BMP-2 knockdown in metastatic breast cancer cells significantly diminished engraftment and colonization. The results suggest metastatic tumor cells create a permissive neural niche by steering NPC differentiation toward astrocytes through paracrine BMP-2 signaling.


Subject(s)
Brain Neoplasms/secondary , Breast Neoplasms/pathology , Neural Stem Cells/cytology , Animals , Bone Morphogenetic Protein 2/physiology , Bone Morphogenetic Protein 4/physiology , Cell Differentiation , Cell Line, Tumor , Coculture Techniques , Female , Glial Fibrillary Acidic Protein , Humans , Mice , Nerve Tissue Proteins/analysis , SOXB1 Transcription Factors/genetics
16.
Oncol Lett ; 5(3): 783-786, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23426187

ABSTRACT

The prevalence of hepatitis B and C in patients with glioblastoma multiforme or meningiomas has not been described. These infections are known to modulate the activity of the immune system, which potentially influences the development and course of cancer. We hypothesized that chronic hepatitis infection, which activates the immune system, decreases the risk of brain tumors, particularly those that are highly malignant. We performed a retrospective study to examine the prevalence of hepatitis B and C in patients with meningiomas and glioblastomas. The combined prevalence of hepatitis B and C in the USA from 1999-2008 was 5.7%. The prevalence of hepatitis B and C in patients with meningiomas was 2.4%; while among glioblastoma patients, the prevalence of hepatitis B and C was 1.38%. The odds ratio of having hepatitis B or C with glioblastoma versus meningiomas was 0.56, with a confidence interval of 0.19-1.6 and a P-value of 0.29. Compared with historical controls, the prevalence of hepatitis B and C in meningioma and glioblastoma patients was decreased. However, this difference may be attributed to the retrospective nature of our data and the natural history of hepatitis B and C infections. The prevalence of these viral infections was not statistically different in patients with meningiomas and glioblastomas. This suggests that hepatitis B and C primarily influence slow-growing, benign tumors and more aggressive cancers equally, if at all. To definitively test our hypothesis, future studies in which data are prospectively gathered are likely to be required.

17.
Neurosurgery ; 72(2): 300-8; discussion 308-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23149951

ABSTRACT

BACKGROUND: Spinal metastases of the second cervical vertebra are a subset of tumors that are particularly difficult to address surgically. Previously described techniques require highly morbid circumferential dissection posterior to the pharynx for resection and reconstruction. OBJECTIVE: To perform a biomechanical analysis of instrumented reconstruction configurations used after axial spondylectomy and to demonstrate safe use of a novel construct in a patient case report. METHODS: Several different published and novel reconstruction configurations were inserted into 7 occipitocervical spines that underwent axial spondylectomy. A biomechanical analysis of the stiffness of the constructs in flexion and extension, lateral bending, and rotation was performed. A patient then underwent a posterior-only approach for axial spondylectomy and circumferential reconstruction. RESULTS: Biomechanical analysis of different constructs demonstrated that anterior column reconstruction with bilateral cages spanning the C1 lateral mass to the C3 facet in combination with occipitocervical instrumentation was superior in flexion-extension and equivalent in lateral bending and rotation to currently used constructs. The patient in whom this construct was placed via a posterior-only approach for axial spondylectomy and instrumentation remained at neurological baseline and demonstrated no recurrence of local disease or failure of instrumentation to date. CONCLUSION: When C1 lateral mass to C3 facet bilateral cage plus occipitocervical instrumentation is compared with existing anterior and posterior constructs, this novel reconstruction is biomechanically equivalent if not superior in performance. In a patient, the posterior-only approach for C2 spondylectomy with the novel reconstruction was safe and durable and avoided the morbidity of the anterior approach.


Subject(s)
Bone Neoplasms/surgery , Internal Fixators , Plastic Surgery Procedures/methods , Sarcoma, Ewing/surgery , Adult , Aged , Biomechanical Phenomena , Bone Neoplasms/secondary , Cadaver , Decompression, Surgical , Female , Humans , Male , Middle Aged , Sarcoma, Ewing/secondary , Spinal Cord/diagnostic imaging , Spinal Cord/surgery , Tomography, X-Ray Computed
18.
Surg Neurol Int ; 3: 136, 2012.
Article in English | MEDLINE | ID: mdl-23230517

ABSTRACT

BACKGROUND: Spinal metastasis is common and can be associated with considerable morbidity. Vertebral resection and reconstruction have been shown to preserve neurological function and decrease pain. Most commonly, two-stage, combined anterior/posterior approaches are performed to surgically address significant vertebral metastasis. Recently, single-stage posterior approaches for vertebrectomies have been performed more often as a result of advances in instrumentation and anesthesia. The objective is to describe a series of patients with metastatic thoracic spine tumors who were treated using a modified, lateral extracavitary approach for a posterior-only vertebral column resection and expandable cage reconstruction. METHODS: A retrospective analysis of 21 cases and 20 patients was performed. RESULTS: The average estimated blood loss and length of surgery were 1700 ml (range, 200-7600 ml) and 6.8 h (range, 4-9 h), respectively. The mean follow-up was 14 months (range, 4-30 months). One patient had a permanent neurological deficit as a result of a postoperative hematoma. Of the five patients who were unable to walk prior to surgery, two regained the ability to ambulate. The total complication rate was 43% with majority being minor. A total of 94% of patients had durable preservation of the neurological function. CONCLUSION: The posterior approach for vertebral column resection and reconstruction is a viable alternative to the standard combined approach. We demonstrate the feasibility of performing the lateral extracavitary approach through a midline incision from T1 to T12. This less invasive approach continues to evolve as instrumentation becomes more advanced and possesses significant advantages in the oncologic setting.

20.
Adv Exp Med Biol ; 760: 188-210, 2012.
Article in English | MEDLINE | ID: mdl-23281521

ABSTRACT

Advancement of in vitro osteogenesis, or the production of bone, is a complex process that has significant clinical implications. Surgical intervention of several spinal disorders entails decompression of the spinal cord and nerves which can lead to subsequent biomechanical instability of the spine. Spinal arthrodesis (fusion) is often required to correct this instability and necessary to eliminate the resulting pathological motion of vertebral segments. Therefore, the achievement of proper spinal fusion, is a critical determinant of treatment efficacy. This chapter focuses on the molecular and cellular components that are involved in bone growth and healing. Mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs) are the precursor cells essential for the formation of the five different types of bone cells: osteoprogenitor cells, osteoblasts, osteoclasts, osteocytes and lining cells. Similarly, endothelial progenitor cells (EPCs) differentiate into endothelial cells, which are essential in angiogenesis and neovascularization. MSCs tri-lineage potential (osteogenic, chondrogenic and adipogenic lineages) have made them the focus of most experimental approaches. Here, we describe their individual roles, as well as pose novel concepts on how their collective role may be the optimal strategy to improve upon in vitro osteogenesis and whether this could also be translated to improved bone formation in vivo. Further, we discuss the various molecular markers that are available for cell identification and the tissue engineering strategies that could replicate the osteoinductive, osteoconductive and osteoproductive milieuthat is available in autograft. Finally, we present a broad primer on the possible integration of cellular, molecular and tissue engineering strategies to improve osteogenesis and the future trends that may bring the promise seen in the laboratory to fruition in preclinical animal models.


Subject(s)
Osteogenesis/physiology , Spinal Diseases/therapy , Spinal Fusion/trends , Tissue Engineering/trends , Animals , Bone Transplantation/methods , Bone Transplantation/trends , Humans , Osteoblasts/cytology , Osteoblasts/physiology , Osteoclasts/cytology , Osteoclasts/physiology , Spinal Diseases/surgery , Spinal Fusion/methods , Spine/embryology , Spine/physiology , Stem Cell Transplantation/methods , Stem Cell Transplantation/trends , Stem Cells/cytology , Stem Cells/physiology , Tissue Engineering/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...