103 results on '"Muldoon LL"'
Search Results
2. Comparison of dynamic susceptibility contrast (DSC) using gadolinium and iron-based contrast agents in high-grade glioma at high-field MRI.
- Author
-
Wongsawaeng D, Schwartz D, Li X, Muldoon LL, Stoller J, Stateler C, Holland S, Szidonya L, Rooney WD, Wyatt C, Ambady P, Fu R, Neuwelt EA, and Barajas RF Jr
- Subjects
- Humans, Male, Female, Middle Aged, Adult, Aged, Gadolinium, Contrast Media, Glioma diagnostic imaging, Magnetic Resonance Imaging methods, Brain Neoplasms diagnostic imaging, Ferrosoferric Oxide
- Abstract
Purpose: To compare DSC-MRI using Gadolinium (GBCA) and Ferumoxytol (FBCA) in high-grade glioma at 3T and 7T MRI field strengths. We hypothesized that using FBCA at 7T would enhance the performance of DSC, as measured by contrast-to-noise ratio (CNR)., Methods: Ten patients (13 lesions) were assigned to 3T (6 patients, 6 lesions) or 7T (4 patients, 7 lesions). All lesions received 0.1 mmol/kg of GBCA on day 1. Ten lesions (4 at 3T and 6 at 7T) received a lower dose (0.6 mg/kg) of FBCA, followed by a higher dose (1.0-1.2 mg/kg), while 3 lesions (2 at 3T and 1 at 7T) received only a higher dose on Day 2. CBV maps with leakage correction for GBCA but not for FBCA were generated. The CNR and normalized CBV (nCBV) were analyzed on enhancing and non-enhancing high T2W lesions., Results: Regardless of FBCA dose, GBCA showed higher CNR than FBCA at 7T, which was significant for high-dose FBCA ( p < .05). Comparable CNR between GBCA and high-dose FBCA was observed at 3T. There was a trend toward higher CNR for FBCA at 3T than 7T. GBCA also showed nCBV twice that of FBCA at both MRI field strengths with significance at 7T., Conclusion: GBCA demonstrated higher image conspicuity, as measured by CNR, than FBCA on 7T. The stronger T2* weighting realized with higher magnetic field strength, combined with FBCA, likely results in more signal loss rather than enhanced performance on DSC. However, at clinical 3T, both GBCA and FBCA, particularly a dosage of 1.0-1.2 mg/kg (optimal for perfusion imaging), yielded comparable CNR., Competing Interests: Declaration of conflicting interestThe author(s) declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article: reports article publishing charges were provided by National Institutes of Health (NIH) grant [K08CA237809], and financial support was provided by The Jonathan D. Lewis Foundation. reports financial support was provided by National Institutes of Health (NIH) grant [NS53468], the Walter S. and Lucienne Driskill Foundation, and AMAG Pharmaceuticals Inc. The other author(s) received no financial support for the research, authorship, and/or publication of this article.
- Published
- 2024
- Full Text
- View/download PDF
3. Deciphering spatially distinct immune microenvironments in glioblastoma using ferumoxytol and gadolinium-enhanced and FLAIR hyperintense MRI phenotypes.
- Author
-
Stoller J, Kersch CN, Muldoon LL, Ambady P, Harrington CA, Fu R, Raslan AM, Dogan A, Neuwelt EA, and Barajas RF Jr
- Abstract
Background: MRI with gadolinium (Gd)-contrast agents is used to assess glioblastoma treatment response but does not specifically reveal heterogeneous biology or immune microenvironmental composition. Ferumoxytol (Fe) contrast is an iron nanoparticle that localizes glioblastoma macrophages and microglia. Therefore, we hypothesized that the use of Fe contrast improves upon standard Gd-based T1-weighted and T2/FLAIR analysis by specifically delineating immune processes., Methods: In this, HIPAA-compliant institutional review board-approved prospective study, stereotactic biopsy samples were acquired from patients with treatment-naïve and recurrent glioblastoma based on MR imaging phenotypes; Gd and Fe T1 enhancement (Gd+, Fe+) or not (Gd-, Fe-), as well as T2-Flair hyperintensity (FLAIR+, FLAIR-). Analysis of genetic expression was performed with RNA microarrays. Imaging and genomic expression patterns were compared using false discovery rate statistics., Results: MR imaging phenotypes defined a variety of immune pathways and Hallmark gene sets. Gene set enrichment analysis demonstrated that Gd+, Fe+, and FLAIR+ features were individually correlated with the same 7 immune process gene sets. Fe+ tissue showed the greatest degree of immune Hallmark gene sets compared to Gd+ or Flair+ tissues and had statistically elevated M2 polarized macrophages, among others. Importantly, the FLAIR+ Gd+ and Fe- imaging phenotypes did not demonstrate expression of immune Hallmark gene sets., Conclusions: Our study demonstrates the potential of Fe and Gd-enhanced MRI phenotypes to reveal spatially distinct immune processes within glioblastoma. Fe improves upon the standard of care Gd enhancement by specifically localizing glioblastoma-associated inflammatory processes, providing valuable insights into tumor biology., Competing Interests: Neither any authors nor their immediate family have a financial relationship with a commercial organization that may have a direct or indirect interest in this content., (© The Author(s) 2023. Published by Oxford University Press, the Society for Neuro-Oncology and the European Association of Neuro-Oncology.)
- Published
- 2023
- Full Text
- View/download PDF
4. Intravenous N-Acetylcysteine to Prevent Cisplatin-Induced Hearing Loss in Children: A Nonrandomized Controlled Phase I Trial.
- Author
-
Orgel E, Knight KR, Chi YY, Malvar J, Rushing T, Mena V, Eisenberg LS, Rassekh SR, Ross CJD, Scott EN, Neely M, Neuwelt EA, Muldoon LL, and Freyer DR
- Subjects
- Adolescent, Humans, Child, Cisplatin adverse effects, Acetylcysteine therapeutic use, Acetylcysteine adverse effects, Administration, Intravenous, Hearing Loss chemically induced, Hearing Loss prevention & control, Neoplasms drug therapy, Neoplasms chemically induced
- Abstract
Purpose: Cisplatin-induced hearing loss (CIHL) is common and permanent. As compared with earlier otoprotectants, we hypothesized N-acetylcysteine (NAC) offers potential for stronger otoprotection through stimulation of glutathione (GSH) production. This study tested the optimal dose, safety, and efficacy of NAC to prevent CIHL., Patients and Methods: In this nonrandomized, controlled phase Ia/Ib trial, children and adolescents newly diagnosed with nonmetastatic, cisplatin-treated tumors received NAC intravenously 4 hours post-cisplatin. The trial performed dose-escalation across three dose levels to establish a safe dose that exceeded the targeted peak serum NAC concentration of 1.5 mmol/L (as identified from preclinical models). Patients with metastatic disease or who were otherwise ineligible were enrolled in an observation-only/control arm. To evaluate efficacy, serial age-appropriate audiology assessments were performed. Integrated biology examined genes involved in GSH metabolism and post-NAC GSH concentrations., Results: Of 52 patients enrolled, 24 received NAC and 28 were in the control arm. The maximum tolerated dose was not reached; analysis of peak NAC concentration identified 450 mg/kg as the recommended phase II dose (RP2D). Infusion-related reactions were common. No severe adverse events occurred. Compared with the control arm, NAC decreased likelihood of CIHL at the end of cisplatin therapy [OR, 0.13; 95% confidence interval (CI), 0.021-0.847; P = 0.033] and recommendations for hearing intervention at end of study (OR, 0.082; 95% CI, 0.011-0.60; P = 0.014). NAC increased GSH; GSTP1 influenced risk for CIHL and NAC otoprotection., Conclusions: NAC was safe at the RP2D, with strong evidence for efficacy to prevent CIHL, warranting further development as a next-generation otoprotectant., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
5. Multiparametric magnetic resonance imaging discerns glioblastoma immune microenvironmental heterogeneity.
- Author
-
Kersch CN, Muldoon LL, Claunch CJ, Fu R, Schwartz D, Cha S, Starkey J, Neuwelt EA, and Barajas RF Jr
- Abstract
Rationale and Objective: Poor clinical outcomes for patients with glioblastoma are in part due to dysfunction of the tumor-immune microenvironment. An imaging approach able to characterize immune microenvironmental signatures could provide a framework for biologically based patient stratification and response assessment. We hypothesized spatially distinct gene expression networks can be distinguished by multiparametric Magnetic Resonance Imaging (MRI) phenotypes., Materials and Methods: Patients with newly diagnosed glioblastoma underwent image-guided tissue sampling allowing for co-registration of MRI metrics with gene expression profiles. MRI phenotypes based on gadolinium contrast enhancing lesion (CEL) and non-enhancing lesion (NCEL) regions were subdivided based on imaging parameters (relative cerebral blood volume (rCBV) and apparent diffusion coefficient (ADC)). Gene set enrichment analysis and immune cell type abundance was estimated using CIBERSORT methodology. Significance thresholds were set at a p -value cutoff 0.005 and an FDR q-value cutoff of 0.1., Results: Thirteen patients (eight men, five women, mean age 58 ± 11 years) provided 30 tissue samples (16 CEL and 14 NCEL). Six non-neoplastic gliosis samples differentiated astrocyte repair from tumor associated gene expression. MRI phenotypes displayed extensive transcriptional variance reflecting biological networks, including multiple immune pathways. CEL regions demonstrated higher immunologic signature expression than NCEL, while NCEL regions demonstrated stronger immune signature expression levels than gliotic non-tumor brain. Incorporation of rCBV and ADC metrics identified sample clusters with differing immune microenvironmental signatures., Conclusion: Taken together, our study demonstrates that MRI phenotypes provide an approach for non-invasively characterizing tumoral and immune microenvironmental glioblastoma gene expression networks.
- Published
- 2023
- Full Text
- View/download PDF
6. Safety of intra-arterial chemotherapy with or without osmotic blood-brain barrier disruption for the treatment of patients with brain tumors.
- Author
-
Uluc K, Ambady P, McIntyre MK, Tabb JP, Kersch CN, Nerison CS, Huddleston A, Liu JJ, Dogan A, Priest RA, Fu R, Prola Netto J, Siler DA, Muldoon LL, Gahramanov S, and Neuwelt EA
- Abstract
Background: Intra-arterial administration of chemotherapy with or without osmotic blood-brain barrier disruption enhances delivery of therapeutic agents to brain tumors. The aim of this study is to evaluate the safety of these procedures., Methods: Retrospectively collected data from a prospective database of consecutive patients with primary and metastatic brain tumors who received intra-arterial chemotherapy without osmotic blood-brain barrier disruption (IA) or intra-arterial chemotherapy with osmotic blood-brain barrier disruption (IA/OBBBD) at Oregon Health and Science University (OHSU) between December 1997 and November 2018 is reported. Chemotherapy-related complications are detailed per Common Terminology Criteria for Adverse Events (CTCAE) guidelines. Procedure-related complications are grouped as major and minor., Results: 4939 procedures (1102 IA; 3837 IA/OBBBD) were performed on 436 patients with various pathologies (primary central nervous system lymphoma [26.4%], glioblastoma [18.1%], and oligodendroglioma [14.7%]). Major procedure-related complications (IA: 12, 1%; IA/OBBBD: 27, 0.7%; P = .292) occurred in 39 procedures including 3 arterial dissections requiring intervention, 21 symptomatic strokes, 3 myocardial infarctions, 6 cervical cord injuries, and 6 deaths within 3 days. Minor procedure-related complications occurred in 330 procedures (IA: 41, 3.7%; IA/OBBBD: 289, 7.5%; P = .001). Chemotherapy-related complications with a CTCAE attribution and grade higher than 3 was seen in 359 (82.3%) patients., Conclusions: We provide safety and tolerability data from the largest cohort of consecutive patients who received IA or IA/OBBBD. Our data demonstrate that IA or IA/OBBBD safely enhance drug delivery to brain tumors and brain around the tumor., (© The Author(s) 2022. Published by Oxford University Press, the Society for Neuro-Oncology and the European Association of Neuro-Oncology.)
- Published
- 2022
- Full Text
- View/download PDF
7. Radiation enhances the delivery of antisense oligonucleotides and improves chemo-radiation efficacy in brain tumor xenografts.
- Author
-
Ambady P, Wu YJ, Kersch CN, Walker JM, Holland S, Muldoon LL, and Neuwelt EA
- Subjects
- Animals, Antineoplastic Agents, Alkylating therapeutic use, Cell Line, Tumor, Heterografts, Humans, Morpholinos, O(6)-Methylguanine-DNA Methyltransferase metabolism, Oligonucleotides, Antisense pharmacology, Rats, Rats, Long-Evans, Xenograft Model Antitumor Assays, Brain Neoplasms drug therapy, Brain Neoplasms radiotherapy, Cerebellar Neoplasms, Medulloblastoma
- Abstract
Overexpression of O
6 -methylguanine DNA methyltransferase (MGMT) contributes to resistance to chemo-radiation therapy (CRT) in brain tumors. We previously demonstrated that non-ablative radiation improved delivery of anti-MGMT morpholino oligonucleotides (AMONs) to reduce MGMT levels in subcutaneous tumor xenografts. We evaluate this approach to enhance CRT efficacy in rat brain tumor xenograft models. The impact of radiation on targeted delivery was evaluated using fluorescent oligonucleotides (f-ON). In vitro, f-ON was localized to clathrin-coated vesicles, endosomes, and lysosomes using confocal microscopy in T98G glioma cells. In vivo, fluorescence was detected in pre-radiated, but not non-radiated Long Evans (non-tumor bearing) rat brains. Cranial radiation (2 Gy) followed by AMONs (intravenous, 10.5 mg/kg) reduced MGMT expression by 50% in both orthotopic cerebellar D283 medulloblastoma and intracerebral H460 non-small cell lung carcinoma (NSCLC) xenograft models. To evaluate the efficacy, AMONs concurrent with CRT (2 Gy radiation plus oral 20 mg/kg temozolomide ×4 days) reduced tumor volumes in the medulloblastoma model (p = 0.012), and a similar trend was found in the NSCLC brain metastasis model. We provide proof of concept for the use of non-ablative radiation to guide and enhance the delivery of morpholino oligonucleotides into brain tumor xenograft models to reduce MGMT levels and improve CRT efficacy., (© 2021. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
8. Adenosine A2A Receptor Activation Enhances Blood-Tumor Barrier Permeability in a Rodent Glioma Model.
- Author
-
Vézina A, Manglani M, Morris D, Foster B, McCord M, Song H, Zhang M, Davis D, Zhang W, Bills J, Nagashima K, Shankarappa P, Kindrick J, Walbridge S, Peer CJ, Figg WD, Gilbert MR, McGavern DB, Muldoon LL, and Jackson S
- Subjects
- Animals, Brain Neoplasms mortality, Disease Models, Animal, Female, Glioma mortality, Humans, Mice, Mice, SCID, Rats, Rats, Nude, Survival Analysis, Transfection, Blood-Brain Barrier metabolism, Brain Neoplasms genetics, Glioma genetics, Receptor, Adenosine A2A metabolism
- Abstract
The blood-tumor barrier (BTB) limits the entry of effective chemotherapeutic agents into the brain for treatment of malignant tumors like glioblastoma. Poor drug entry across the BTB allows infiltrative glioma stem cells to evade therapy and develop treatment resistance. Regadenoson, an FDA-approved adenosine A2A receptor (A2AR) agonist, has been shown to increase drug delivery across the blood-brain barrier in non-tumor-bearing rodents without a defined mechanism of enhancing BTB permeability. Here, we characterize the time-dependent impact of regadenoson on brain endothelial cell interactions and paracellular transport, using mouse and rat brain endothelial cells and tumor models. In vitro , A2AR activation leads to disorganization of cytoskeletal actin filaments by 30 minutes, downregulation of junctional protein expression by 4 hours, and reestablishment of endothelial cell integrity by 8 hours. In rats bearing intracranial gliomas, regadenoson treatment results in increase of intratumoral temozolomide concentrations, yet no increased survival noted with combined temozolomide therapy. These findings demonstrate regadenoson's ability to induce brain endothelial structural changes among glioma to increase BTB permeability. The use of vasoactive mediators, like regadenoson, which transiently influences paracellular transport, should further be explored to evaluate their potential to enhance central nervous system treatment delivery to aggressive brain tumors. IMPLICATIONS: This study provides insight on the use of a vasoactive agent to increase exposure of the BTB to chemotherapy with intention to improve glioma treatment efficacy., (©2021 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
9. Blood-brain barrier opening by intracarotid artery hyperosmolar mannitol induces sterile inflammatory and innate immune responses.
- Author
-
Burks SR, Kersch CN, Witko JA, Pagel MA, Sundby M, Muldoon LL, Neuwelt EA, and Frank JA
- Subjects
- Animals, Blood-Brain Barrier metabolism, Carotid Arteries drug effects, Cell Adhesion drug effects, Cell Adhesion Molecules blood, Central Nervous System Neoplasms drug therapy, Central Nervous System Neoplasms genetics, Chemokines blood, Cytokines blood, Endothelial Cells drug effects, Humans, Inflammation blood, Rats, Tight Junctions drug effects, Tight Junctions genetics, Blood-Brain Barrier drug effects, Immunity, Innate genetics, Inflammation genetics, Mannitol pharmacology
- Abstract
Intracarotid arterial hyperosmolar mannitol (ICAHM) blood-brain barrier disruption (BBBD) is effective and safe for delivery of therapeutics for central nervous system malignancies. ICAHM osmotically alters endothelial cells and tight junction integrity to achieve BBBD. However, occurrence of neuroinflammation following hemispheric BBBD by ICAHM remains unknown. Temporal proteomic changes in rat brains following ICAHM included increased damage-associated molecular patterns, cytokines, chemokines, trophic factors, and cell adhesion molecules, indicative of a sterile inflammatory response (SIR). Proteomic changes occurred within 5 min of ICAHM infusion and returned to baseline by 96 h. Transcriptomic analyses following ICAHM BBBD further supported an SIR. Immunohistochemistry revealed activated astrocytes, microglia, and macrophages. Moreover, proinflammatory proteins were elevated in serum, and proteomic and histological findings from the contralateral hemisphere demonstrated a less pronounced SIR, suggesting neuroinflammation beyond regions of ICAHM infusion. Collectively, these results demonstrate ICAHM induces a transient SIR that could potentially be harnessed for neuroimmunomodulation., Competing Interests: The authors declare no competing interest.
- Published
- 2021
- Full Text
- View/download PDF
10. High dose acetaminophen inhibits STAT3 and has free radical independent anti-cancer stem cell activity.
- Author
-
Pingali P, Wu YJ, Boothello R, Sharon C, Li H, Sistla S, Sankaranarayanan NV, Desai UR, Le AT, Doebele RC, Muldoon LL, Patel BB, and Neuwelt A
- Subjects
- AC133 Antigen metabolism, Acetaminophen administration & dosage, Antineoplastic Agents administration & dosage, Biomarkers, Tumor, Cell Line, Tumor, Cell Proliferation drug effects, Dose-Response Relationship, Drug, Gene Knockdown Techniques, Humans, Interleukin-6 antagonists & inhibitors, Lung Neoplasms, STAT3 Transcription Factor genetics, STAT3 Transcription Factor metabolism, Acetaminophen pharmacology, Antineoplastic Agents pharmacology, Free Radicals metabolism, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells metabolism, STAT3 Transcription Factor antagonists & inhibitors
- Abstract
High-dose acetaminophen (AAP) with delayed rescue using n-acetylcysteine (NAC), the FDA-approved antidote to AAP overdose, has demonstrated promising antitumor efficacy in early phase clinical trials. However, the mechanism of action (MOA) of AAP's anticancer effects remains elusive. Using clinically relevant AAP concentrations, we evaluated cancer stem cell (CSC) phenotype in vitro and in vivo in lung cancer and melanoma cells with diverse driver mutations. Associated mechanisms were also studied. Our results demonstrated that AAP inhibited 3D spheroid formation, self-renewal, and expression of CSC markers when human cancer cells were grown in serum-free CSC media. Similarly, anti-CSC activity was demonstrated in vivo in xenograft models - tumor formation following in vitro treatment and ex-vivo spheroid formation following in vivo treatment. Intriguingly, NAC, used to mitigate AAP's liver toxicity, did not rescue cells from AAP's anti-CSC effects, and AAP failed to reduce glutathione levels in tumor xenograft in contrast to mice liver tissue suggesting nonglutathione-related MOA. In fact, AAP mediates its anti-CSC effect via inhibition of STAT3. AAP directly binds to STAT3 with an affinity in the low micromolar range and a high degree of specificity for STAT3 relative to STAT1. These findings have high immediate translational significance concerning advancing AAP with NAC rescue to selectively rescue hepatotoxicity while inhibiting CSCs. The novel mechanism of selective STAT3 inhibition has implications for developing rational anticancer combinations and better patient selection (predictive biomarkers) for clinical studies and developing novel selective STAT3 inhibitors using AAP's molecular scaffold., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
11. Transcriptional signatures in histologic structures within glioblastoma tumors may predict personalized drug sensitivity and survival.
- Author
-
Kersch CN, Claunch CJ, Ambady P, Bucher E, Schwartz DL, Barajas RF Jr, Iliff JJ, Risom T, Heiser L, Muldoon LL, Korkola JE, Gray JW, and Neuwelt EA
- Abstract
Background: Glioblastoma is a rapidly fatal brain cancer that exhibits extensive intra- and intertumoral heterogeneity. Improving survival will require the development of personalized treatment strategies that can stratify tumors into subtypes that differ in therapeutic vulnerability and outcomes. Glioblastoma stratification has been hampered by intratumoral heterogeneity, limiting our ability to compare tumors in a consistent manner. Here, we develop methods that mitigate the impact of intratumoral heterogeneity on transcriptomic-based patient stratification., Methods: We accessed open-source transcriptional profiles of histological structures from 34 human glioblastomas from the Ivy Glioblastoma Atlas Project. Principal component and correlation network analyses were performed to assess sample inter-relationships. Gene set enrichment analysis was used to identify enriched biological processes and classify glioblastoma subtype. For survival models, Cox proportional hazards regression was utilized. Transcriptional profiles from 156 human glioblastomas were accessed from The Cancer Genome Atlas to externally validate the survival model., Results: We showed that intratumoral histologic architecture influences tumor classification when assessing established subtyping and prognostic gene signatures, and that indiscriminate sampling can produce misleading results. We identified the cellular tumor as a glioblastoma structure that can be targeted for transcriptional analysis to more accurately stratify patients by subtype and prognosis. Based on expression from cellular tumor, we created an improved risk stratification gene signature., Conclusions: Our results highlight that biomarker performance for diagnostics, prognostics, and prediction of therapeutic response can be improved by analyzing transcriptional profiles in pure cellular tumor, which is a critical step toward developing personalized treatment for glioblastoma., (© The Author(s) 2020. Published by Oxford University Press, the Society for Neuro-Oncology and the European Association of Neuro-Oncology.)
- Published
- 2020
- Full Text
- View/download PDF
12. Distinguishing Extravascular from Intravascular Ferumoxytol Pools within the Brain: Proof of Concept in Patients with Treated Glioblastoma.
- Author
-
Barajas RF Jr, Schwartz D, McConnell HL, Kersch CN, Li X, Hamilton BE, Starkey J, Pettersson DR, Nickerson JP, Pollock JM, Fu RF, Horvath A, Szidonya L, Varallyay CG, Jaboin JJ, Raslan AM, Dogan A, Cetas JS, Ciporen J, Han SJ, Ambady P, Muldoon LL, Woltjer R, Rooney WD, and Neuwelt EA
- Subjects
- Adult, Artifacts, Contrast Media analysis, Contrast Media metabolism, Female, Ferrosoferric Oxide metabolism, Humans, Macrophages metabolism, Male, Middle Aged, Neuroimaging methods, Proof of Concept Study, Brain Neoplasms diagnostic imaging, Ferrosoferric Oxide analysis, Glioblastoma diagnostic imaging, Image Interpretation, Computer-Assisted methods, Magnetic Resonance Imaging methods
- Abstract
Background and Purpose: Glioblastoma-associated macrophages are a major constituent of the immune response to therapy and are known to engulf the iron-based MR imaging contrast agent, ferumoxytol. Current ferumoxytol MR imaging techniques for localizing macrophages are confounded by contaminating intravascular signal. The aim of this study was to assess the utility of a newly developed MR imaging technique, segregation and extravascular localization of ferumoxytol imaging, for differentiating extravascular-from-intravascular ferumoxytol contrast signal at a delayed 24-hour imaging time point., Materials and Methods: Twenty-three patients with suspected post-chemoradiotherapy glioblastoma progression underwent ferumoxytol-enhanced SWI. Segregation and extravascular localization of ferumoxytol imaging maps were generated as the voxelwise difference of the delayed (24 hours) from the early (immediately after administration) time point SWI maps. Continuous segregation and extravascular localization of ferumoxytol imaging map values were separated into positive and negative components. Image-guided biologic correlation was performed., Results: Negative segregation and extravascular localization of ferumoxytol imaging values correlated with early and delayed time point SWI values, demonstrating that intravascular signal detected in the early time point persists into the delayed time point. Positive segregation and extravascular localization of ferumoxytol imaging values correlated only with delayed time point SWI values, suggesting successful detection of the newly developed extravascular signal., Conclusions: Segregation and extravascular localization of ferumoxytol MR imaging improves on current techniques by eliminating intrinsic tissue and intravascular ferumoxytol signal and may inform glioblastoma outcomes by serving as a more specific metric of macrophage content compared with uncorrected T1 and SWI techniques., (© 2020 by American Journal of Neuroradiology.)
- Published
- 2020
- Full Text
- View/download PDF
13. Toll-like Receptor 4 Signaling and Downstream Neutrophilic Inflammation Mediate Endotoxemia-Enhanced Blood-Labyrinth Barrier Trafficking.
- Author
-
Urdang ZD, Bills JL, Cahana DY, Muldoon LL, and Neuwelt EA
- Subjects
- Animals, Chemotaxis, Leukocyte immunology, Inflammation chemically induced, Inflammation immunology, Lipopolysaccharides toxicity, Mice, Mice, Knockout, Neutrophils immunology, Ototoxicity immunology, Cytokines immunology, Ear, Inner immunology, Endotoxemia immunology, Neutrophil Infiltration immunology, Signal Transduction immunology, Toll-Like Receptor 4 immunology
- Abstract
Hypothesis: Both toll-like receptor 4 (TLR4) and downstream neutrophil activity are required for endotoxemia-enhanced blood-labyrinth barrier (BLB) trafficking., Background: Aminoglycoside and cisplatin are valuable clinical therapies; however, these drugs often cause life-long hearing loss. Endotoxemia enhances the ototoxicity of aminoglycosides and cisplatin in a TLR4 dependent mechanism for which downstream proinflammatory signaling orchestrates effector immune cells including neutrophils. Neutrophil-mediated vascular injury (NMVI) can enhance molecular trafficking across endothelial barriers and may contribute to endotoxemia-enhanced drug-induced ototoxicity., Methods: Lipopolysaccharide (LPS) hypo-responsive TLR4-KO mice and congenitally neutropenic granulocyte colony-stimulating factor (GCSF) GCSF-KO mice were studied to investigate the relative contributions of TLR4 signaling and downstream neutrophil activity to endotoxemia-enhanced BLB trafficking. C57Bl/6 wild-type mice were used as a positive control. Mice were treated with LPS and 24 hours later cochleae were analyzed for gene transcription of innate inflammatory cytokine/chemokine signaling molecules, neutrophil recruitment, and vascular trafficking of the paracellular tracer biocytin-TMR., Results: Cochlear transcription of innate proinflammatory cytokines/chemokines was increased in endotoxemic C57Bl/6 and GCSF-KO, but not in TLR4-KO mice. More neutrophils were recruited to endotoxemic C57Bl/6 cochleae compared with both TLR4 and GCSF-KO cochleae. Endotoxemia enhanced BLB trafficking of biocytin-TMR in endotoxemic C57Bl/6 cochleae and this was attenuated in both TLR4 and GCSF-KO mice., Conclusion: Together these results suggest that TLR4-mediated innate immunity cytokine/chemokine signaling alone is not sufficient for endotoxemia-enhanced trafficking of biocytin-TMR and that downstream neutrophil activity is required to enhance BLB trafficking. Clinically, targeting neutrophilic inflammation could protect hearing during aminoglycoside, cisplatin, or other ototoxic drug therapies.
- Published
- 2020
- Full Text
- View/download PDF
14. Combined iron oxide nanoparticle ferumoxytol and gadolinium contrast enhanced MRI define glioblastoma pseudoprogression.
- Author
-
Barajas RF, Hamilton BE, Schwartz D, McConnell HL, Pettersson DR, Horvath A, Szidonya L, Varallyay CG, Firkins J, Jaboin JJ, Kubicky CD, Raslan AM, Dogan A, Cetas JS, Ciporen J, Han SJ, Ambady P, Muldoon LL, Woltjer R, Rooney WD, and Neuwelt EA
- Subjects
- Adult, Brain Neoplasms pathology, Contrast Media, Female, Glioblastoma pathology, Humans, Magnetite Nanoparticles, Male, Middle Aged, Neoplasm Recurrence, Local diagnostic imaging, Neoplasm Recurrence, Local pathology, Retrospective Studies, Brain Neoplasms diagnostic imaging, Ferrosoferric Oxide, Gadolinium, Glioblastoma diagnostic imaging, Magnetic Resonance Imaging methods
- Abstract
Background: Noninvasively differentiating therapy-induced pseudoprogression from recurrent disease in patients with glioblastoma is prospectively difficult due to the current lack of a biologically specific imaging metric. Ferumoxytol iron oxide nanoparticle MRI contrast characterizes innate immunity mediated neuroinflammation; therefore, we hypothesized that combined ferumoxytol and gadolinium enhanced MRI could serve as a biomarker of glioblastoma pseudoprogression., Methods: In this institutional review board-approved, retrospective study, we analyzed ferumoxytol and gadolinium contrast enhanced T1-weighted 3T MRI in 45 patients with glioblastoma over multiple clinical timepoints. Isocitrate dehydrogenase 1 (IDH-1) mutational status was characterized by exome sequencing. Sum of products diameter measurements were calculated according to Response Assessment in Neuro-Oncology criteria from both gadolinium and ferumoxytol enhanced sequences. Enhancement mismatch was calculated as the natural log of the ferumoxytol to gadolinium sum of products diameter ratio. Analysis of variance and Student's t-test assessed differences in mismatch ratios. P-value <0.05 indicated statistical significance., Results: With the development of pseudoprogression we observed a significantly elevated mismatch ratio compared with disease recurrence (P < 0.01) within IDH-1 wild type patients. Patients with IDH-1 mutation demonstrated significantly reduced mismatch ratio with the development of pseudoprogression compared with disease recurrence (P < 0.01). Receiver operator curve analysis demonstrated 100% sensitivity and specificity for the use of mismatch ratios as a diagnostic biomarker of pseudoprogression., Conclusion: Our study suggests that ferumoxytol to gadolinium contrast mismatch ratios are an MRI biomarker for the diagnosis of pseudoprogression in patients with glioblastoma. This may be due to the unique characterization of therapy-induced neuroinflammation., (Published by Oxford University Press on behalf of the Society for Neuro-Oncology 2018.)
- Published
- 2019
- Full Text
- View/download PDF
15. Cerebral blood volume mapping with ferumoxytol in dynamic susceptibility contrast perfusion MRI: Comparison to standard of care.
- Author
-
Varallyay CG, Nesbit E, Horvath A, Varallyay P, Fu R, Gahramanov S, Muldoon LL, Li X, Rooney WD, and Neuwelt EA
- Subjects
- Adult, Aged, Brain Mapping, Contrast Media, Female, Gadolinium chemistry, Humans, Male, Metal Nanoparticles, Middle Aged, Perfusion, Retrospective Studies, Cerebrovascular Circulation, Ferric Compounds chemistry, Ferrosoferric Oxide chemistry, Heterocyclic Compounds chemistry, Magnetic Resonance Imaging, Organometallic Compounds chemistry
- Abstract
Background: Cerebral blood volume (CBV) mapping with a dynamic susceptibility contrast (DSC) perfusion technique has become a clinical tool in diagnosing and follow-up of brain tumors. Ferumoxytol, a long-circulating iron oxide nanoparticle, has been tested for CBV mapping, but the optimal dose has not been established., Purpose: To compare ferumoxytol DSC of two different doses to standard of care gadoteridol by analyzing time-intensity curves and CBV maps in normal-appearing brain regions., Study Type: Retrospective., Subjects: Fifty-four patients with various brain disorders., Field Strength/sequence: 3T MRI. DSC-MRI was performed with 0.1 mmol/kg gadoteridol and 1 day later with ferumoxytol in doses of 1 or 2 mg/kg., Assessment: Signal changes during first pass, relative CBV (rCBV) in normal-appearing thalamus, putamen, and globus pallidus, and contrast-to-noise ratio (CNR) of the CBV maps were compared between gadoteridol and various doses of ferumoxytol using an automated method. To subjectively assess the quality of the CBV maps, two blinded readers also assessed visual conspicuity of the putamen., Statistical Tests: Linear mixed effect model was used for statistical comparison., Results: Compared to gadoteridol, 1 mg/kg ferumoxytol showed no difference in CNR (P = 0.6505), peak ΔR2*, and rCBV in the putamen (P = 0.2669, 0.0871) or in the thalamus (P = 0.517, 0.9787); 2 mg/kg ferumoxytol increased peak ΔR2* as well as the CNR (P < 0.0001), but also mildly increased rCBV in putamen and globus pallidus (P = 0.0005, 0.0012). Signal intensities during first pass remained highly above the noise level, with overlapping of 95% confidence intervals with noise only in 3 out of 162 tested regions. Compared to gadoteridol, the visual image quality showed mild improvement with 1 mg/kg (P = 0.02) and marked improvement with 2 mg/kg ferumoxytol (P < 0.0001)., Data Conclusion: 1 mg/kg ferumoxytol provides similar imaging results to standard gadoteridol for DSC-MRI, and 2 mg/kg has a benefit of increased CNR, but may also result in mildly increased rCBV values., Level of Evidence: 3 Technical Efficacy: Stage 1 J. MAGN. RESON. IMAGING 2018;48:441-448., (© 2018 International Society for Magnetic Resonance in Medicine.)
- Published
- 2018
- Full Text
- View/download PDF
16. Enhancing the cytotoxicity of chemoradiation with radiation-guided delivery of anti-MGMT morpholino oligonucleotides in non-methylated solid tumors.
- Author
-
Ambady P, Wu YJ, Walker JM, Kersch C, Pagel MA, Woltjer RL, Fu R, Muldoon LL, and Neuwelt EA
- Subjects
- A549 Cells, Animals, Apoptosis drug effects, Apoptosis genetics, Apoptosis radiation effects, Cell Line, Tumor, Chemoradiotherapy, DNA Modification Methylases biosynthesis, DNA Repair Enzymes biosynthesis, Female, Humans, Immunohistochemistry, Morpholinos administration & dosage, Morpholinos genetics, Morpholinos pharmacokinetics, Neoplasms genetics, Neoplasms metabolism, Neoplasms radiotherapy, Oligonucleotides, Antisense genetics, Oligonucleotides, Antisense pharmacokinetics, Rats, Rats, Nude, Transfection, Tumor Suppressor Proteins biosynthesis, DNA Modification Methylases genetics, DNA Repair Enzymes genetics, Neoplasms therapy, Oligonucleotides, Antisense administration & dosage, Tumor Suppressor Proteins genetics
- Abstract
The DNA repair enzyme O
6 -methylguanine DNA methyltransferase (MGMT) is epigenetically silenced in some tumors by MGMT gene promoter methylation. MGMT-hypermethylated solid tumors have enhanced susceptibility to the cytotoxic effects of alkylating chemotherapy such as temozolomide, compared with non-methylated tumors. In glioblastoma, subjects with MGMT hypermethylation have significantly longer survival rates after chemoradiotherapy. We report the first successful use of a non-ablative dose of ionizing radiation to prime human cancer cells to enhance the uptake of unmodified anti-MGMT morpholino oligonucleotide (AMON) sequences. We demonstrate >40% reduction in the in vitro proliferation index and cell viability in radiation-primed MGMT-expressing human solid tumor cells treated with a single dose of AMONs and temozolomide. We further demonstrate the feasibility of using a non-ablative dose of radiation in vivo to guide and enhance the delivery of intravenously administered AMONs to achieve 50% MGMT knockdown only at radiation-primed tumor sites in a subcutaneous tumor model. Local upregulation of physiological endocytosis after radiation may have a role in radiation-guided uptake of AMONs. This approach holds direct translational significance in glioblastoma and brain metastases where radiation is part of the standard of care; our approach to silence MGMT could overcome the significant problem of MGMT-mediated chemoresistance.- Published
- 2017
- Full Text
- View/download PDF
17. High αv Integrin Level of Cancer Cells Is Associated with Development of Brain Metastasis in Athymic Rats.
- Author
-
Wu YJ, Pagel MA, Muldoon LL, Fu R, and Neuwelt EA
- Subjects
- Animals, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal, Humanized, Brain metabolism, Brain pathology, Brain Neoplasms drug therapy, Brain Neoplasms pathology, Brain Neoplasms secondary, Disease Models, Animal, Gene Expression Regulation, Neoplastic genetics, Humans, Integrin alphaV biosynthesis, Neoplasm Metastasis, Neoplasms drug therapy, Neoplasms pathology, Rats, Brain Neoplasms genetics, Cell Adhesion genetics, Integrin alphaV genetics, Neoplasms genetics
- Abstract
Background/aim: Brain metastases commonly occur in patients with malignant skin, lung and breast cancers resulting in high morbidity and poor prognosis. Integrins containing an αv subunit are cell adhesion proteins that contribute to cancer cell migration and cancer progression. We hypothesized that high expression of αv integrin cell adhesion protein promoted metastatic phenotypes in cancer cells., Materials and Methods: Cancer cells from different origins were used and studied regarding their metastatic ability and intetumumab, anti-αv integrin mAb, sensitivity using in vitro cell migration assay and in vivo brain metastases animal models., Results: The number of brain metastases and the rate of occurrence were positively correlated with cancer cell αv integrin levels. High αv integrin-expressing cancer cells showed significantly faster cell migration rate in vitro than low αv integrin-expressing cells. Intetumumab significantly inhibited cancer cell migration in vitro regardless of αv integrin expression level. Overexpression of αv integrin in cancer cells with low αv integrin level accelerated cell migration in vitro and increased the occurrence of brain metastases in vivo., Conclusion: αv integrin promotes brain metastases in cancer cells and may mediate early steps in the metastatic cascade, such as adhesion to brain vasculature. Targeting αv integrin with intetumumab could provide clinical benefit in treating cancer patients who develop metastases., (Copyright© 2017, International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
18. Current and potential imaging applications of ferumoxytol for magnetic resonance imaging.
- Author
-
Toth GB, Varallyay CG, Horvath A, Bashir MR, Choyke PL, Daldrup-Link HE, Dosa E, Finn JP, Gahramanov S, Harisinghani M, Macdougall I, Neuwelt A, Vasanawala SS, Ambady P, Barajas R, Cetas JS, Ciporen J, DeLoughery TJ, Doolittle ND, Fu R, Grinstead J, Guimaraes AR, Hamilton BE, Li X, McConnell HL, Muldoon LL, Nesbit G, Netto JP, Petterson D, Rooney WD, Schwartz D, Szidonya L, and Neuwelt EA
- Subjects
- Adolescent, Adult, Animals, Atlases as Topic, Child, Preschool, Contrast Media adverse effects, Contrast Media pharmacokinetics, Female, Ferrosoferric Oxide adverse effects, Ferrosoferric Oxide pharmacokinetics, Hematinics administration & dosage, Humans, Kidney physiopathology, Magnetic Resonance Imaging adverse effects, Male, Middle Aged, Predictive Value of Tests, Renal Elimination, Renal Insufficiency, Chronic physiopathology, Reproducibility of Results, Contrast Media administration & dosage, Ferrosoferric Oxide administration & dosage, Magnetic Resonance Imaging methods
- Abstract
Contrast-enhanced magnetic resonance imaging is a commonly used diagnostic tool. Compared with standard gadolinium-based contrast agents, ferumoxytol (Feraheme, AMAG Pharmaceuticals, Waltham, MA), used as an alternative contrast medium, is feasible in patients with impaired renal function. Other attractive imaging features of i.v. ferumoxytol include a prolonged blood pool phase and delayed intracellular uptake. With its unique pharmacologic, metabolic, and imaging properties, ferumoxytol may play a crucial role in future magnetic resonance imaging of the central nervous system, various organs outside the central nervous system, and the cardiovascular system. Preclinical and clinical studies have demonstrated the overall safety and effectiveness of this novel contrast agent, with rarely occurring anaphylactoid reactions. The purpose of this review is to describe the general and organ-specific properties of ferumoxytol, as well as the advantages and potential pitfalls associated with its use in magnetic resonance imaging. To more fully demonstrate the applications of ferumoxytol throughout the body, an imaging atlas was created and is available online as supplementary material., (Published by Elsevier Inc.)
- Published
- 2017
- Full Text
- View/download PDF
19. Ferumoxytol nanoparticle uptake in brain during acute neuroinflammation is cell-specific.
- Author
-
McConnell HL, Schwartz DL, Richardson BE, Woltjer RL, Muldoon LL, and Neuwelt EA
- Subjects
- Animals, Brain Neoplasms diagnostic imaging, Contrast Media, Humans, Rats, Brain pathology, Ferrosoferric Oxide pharmacokinetics, Magnetic Resonance Imaging, Nanoparticles
- Abstract
Ferumoxytol ultrasmall superparamagnetic iron oxide nanoparticles can enhance contrast between neuroinflamed and normal-appearing brain tissue when used as a contrast agent for high-sensitivity magnetic resonance imaging (MRI). Here we used an anti-dextran antibody (Dx1) that binds the nanoparticle's carboxymethyldextran coating to differentiate ferumoxytol from endogenous iron and localize it unequivocally in brain tissue. Intravenous injection of ferumoxytol into immune-competent rats that harbored human tumor xenograft-induced inflammatory brain lesions resulted in heterogeneous and lesion-specific signal enhancement on MRI scans in vivo. We used Dx1 immunolocalization and electron microscopy to identify ferumoxytol in affected tissue post-MRI. We found that ferumoxytol nanoparticles were taken up by astrocyte endfeet surrounding cerebral vessels, astrocyte processes, and CD163(+)/CD68(+) macrophages, but not by tumor cells. These results provide a biological basis for the delayed imaging changes seen with ferumoxytol and indicate that ferumoxytol-MRI can be used to assess the inflammatory component of brain lesions in the clinic., (Published by Elsevier Inc.)
- Published
- 2016
- Full Text
- View/download PDF
20. Intra-arterial administration improves temozolomide delivery and efficacy in a model of intracerebral metastasis, but has unexpected brain toxicity.
- Author
-
Muldoon LL, Pagel MA, Netto JP, and Neuwelt EA
- Subjects
- Animals, Antineoplastic Agents, Alkylating administration & dosage, Antineoplastic Agents, Alkylating adverse effects, Brain Neoplasms secondary, Dacarbazine administration & dosage, Dacarbazine adverse effects, Humans, Injections, Intra-Arterial, Lung Neoplasms pathology, Male, Middle Aged, Neurotoxicity Syndromes pathology, Rats, Rats, Nude, Small Cell Lung Carcinoma pathology, Temozolomide, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Blood-Brain Barrier drug effects, Brain Neoplasms drug therapy, Dacarbazine analogs & derivatives, Drug Delivery Systems, Lung Neoplasms drug therapy, Neurotoxicity Syndromes etiology, Small Cell Lung Carcinoma drug therapy
- Abstract
We tested the hypothesis that intra-arterial (IA) infusion of temozolomide into the internal carotid artery would safely improve drug delivery to brain and enhance chemotherapy efficacy in a chemosensitive rat brain tumor model. Quantitative autoradiography after 25 µCi (14)C-temozolomide was given by oral, intravenous, or IA route of administration, or IA with osmotic blood-brain barrier disruption (BBBD) (n = 5-7 per group) showed that both IA and IA/BBBD administration increased drug delivery in tumor by over threefold compared to normal brain (P < 0.02), and also significantly elevated delivery throughout the infused right hemisphere. Temozolomide (20 mg/kg; ~150 mg/m(2)) increased median survival when given by oral (25.5 days), intravenous (25.5 days), or IA (33 days) route of administration, compared to 17.5 days in untreated controls (n = 8 per group; overall P < 0.0001). Survival time after IA temozolomide was significantly longer than all other groups (P < 0.01 for all comparisons). BBBD temozolomide was toxic in the efficacy study, but there was no evidence of symptomatic neurotoxicity in rats given IA temozolomide. After these promising animal results, a 49 year old male with glioblastoma multiforme who failed all standard therapy received temozolomide 100 mg/m(2) IA. Upon initiation of the second course of IA infusion the patient had increased heart rate, blood pressure, and rash, and the procedure was terminated without sequelae. Follow up IA infusion of temozolomide diluent in normal rats showed damaged cerebrovasculature as determined by dye leakage. These results demonstrate that IA infusion of temozolomide was toxic, with or without BBBD. We conclude that under the current formulation temozolomide is not safe for IA infusion in patients.
- Published
- 2016
- Full Text
- View/download PDF
21. Dose-Dependent Neurotoxicity (Seizures) Due to Deposition of Gadolinium-based Contrast Agents in the Central Nervous System.
- Author
-
Muldoon LL and Neuwelt EA
- Subjects
- Animals, Contrast Media adverse effects, Dogs, Dose-Response Relationship, Drug, Gadolinium adverse effects, Humans, Brain drug effects, Contrast Media administration & dosage, Gadolinium administration & dosage, Seizures chemically induced
- Published
- 2015
- Full Text
- View/download PDF
22. Interactions between αv-Integrin and HER2 and Their Role in the Invasive Phenotype of Breast Cancer Cells In Vitro and in Rat Brain.
- Author
-
Lal S, Kersch C, Beeson KA, Wu YJ, Muldoon LL, and Neuwelt EA
- Subjects
- Animals, Brain pathology, Breast Neoplasms genetics, Breast Neoplasms pathology, Cell Line, Tumor, Female, Gene Knockdown Techniques, Heterografts, Humans, Integrin alphaV genetics, Neoplasm Invasiveness, Neoplasm Transplantation, Rats, Receptor, ErbB-2 genetics, Brain metabolism, Breast Neoplasms metabolism, Integrin alphaV metabolism, Receptor, ErbB-2 metabolism
- Abstract
Background: We tested the hypothesis that αv-integrin and the human epidermal growth factor receptor type 2 (HER2) interact with each other in brain trophic metastatic breast cancer cells and influence their invasive phenotype., Methods: Clones of MDA-MB231BR human breast cancer cells with stable knock down of αv-integrin in combination with high or low levels of HER2 were created. The interactions of these two proteins and their combined effect on cell migration and invasion were investigated in vitro and in vivo., Results: Knockdown of αv-integrin in MDA-MB231BR clones altered the actin cytoskeleton and cell morphology. HER2 co-precipitated with αv-integrin in three breast cancer cell lines in vitro, suggesting they complex in cells. Knockdown of αv-integrin altered HER2 localization from its normal membrane position to a predominantly lysosomal localization. When αv-integrin expression was decreased by 69-93% in HER2-expressing cells, cellular motility was significantly reduced. Deficiency of both αv-integrin and HER2 decreased cellular migration and invasion by almost 90% compared to cells expressing both proteins (P<0.01). After intracerebral inoculation, cells expressing high levels of both αv-integrin and HER2 showed a diffusely infiltrative tumor phenotype, while cells deficient in αv-integrin and/or HER2 showed a compact tumor growth phenotype. In the αv-integrin positive/HER2 positive tumors, infiltrative growth was 57.2 ± 19% of tumor volume, compared to only 5.8 ± 6.1% infiltration in the double deficient tumor cells., Conclusions: αv-integrin interacts with HER2 in breast cancer cells and may regulate HER2 localization. The combined impacts of αv-integrin and HER2 influence the invasive phenotype of breast cancer cells. Targeting αv-integrin in HER2-positive breast cancer may slow growth and decrease infiltration in the normal brain.
- Published
- 2015
- Full Text
- View/download PDF
23. Vascular endothelial growth factor blockade alters magnetic resonance imaging biomarkers of vascular function and decreases barrier permeability in a rat model of lung cancer brain metastasis.
- Author
-
Pishko GL, Muldoon LL, Pagel MA, Schwartz DL, and Neuwelt EA
- Subjects
- Animals, Biomarkers, Brain metabolism, Brain Neoplasms metabolism, Brain Neoplasms pathology, Brain Neoplasms secondary, Capillary Permeability drug effects, Cell Line, Tumor, Disease Models, Animal, Female, Humans, Magnetic Resonance Imaging, Permeability, Rats, Rats, Nude, Treatment Outcome, Adenocarcinoma pathology, Angiogenesis Inhibitors therapeutic use, Bevacizumab therapeutic use, Brain blood supply, Brain Neoplasms drug therapy, Lung Neoplasms pathology, Neovascularization, Pathologic drug therapy
- Abstract
Background: Blockade of vascular endothelial growth factor (VEGF) to promote vascular normalization and inhibit angiogenesis has been proposed for the treatment of brain metastases; however, vascular normalization has not been well-characterized in this disease. We investigated the effect of treatment with bevacizumab anti-VEGF antibody on magnetic resonance imaging (MRI) biomarkers of brain tumor vascular characteristics in comparison to small molecule delivery in a rat model of human lung cancer brain metastasis., Methods: Athymic rats with A549 human lung adenocarcinoma intracerebral xenografts underwent MRI at 11.75 T before and one day after treatment with bevacizumab (n = 8) or saline control (n = 8) to evaluate tumor volume, free water content (edema), blood volume and vascular permeability (Ktrans). One day later, permeability to 14C-aminoisobutyric acid (AIB) was measured in tumor and brain to assess the penetration of a small drug-like molecule., Results: In saline control animals, tumor volume, edema and permeability increased over the two day assessment period. Compared to controls, bevacizumab treatment slowed the rate of tumor growth (P = 0.003) and blocked the increase in edema (P = 0.033), but did not alter tumor blood volume. Bevacizumab also significantly reduced Ktrans (P = 0.033) and AIB passive permeability in tumor (P = 0.04), but not to peritumoral tissue or normal brain. Post-treatment Ktrans correlated with AIB levels in the bevacizumab-treated rats but not in the saline controls., Conclusions: The correlation of an MRI biomarker for decreased vascular permeability with decreased AIB concentration in tumor after antiangiogenic treatment suggests that bevacizumab partially restored the normal low permeability characteristics of the blood-brain barrier in a model of human lung cancer brain metastasis.
- Published
- 2015
- Full Text
- View/download PDF
24. N-acetylcysteine chemoprotection without decreased cisplatin antitumor efficacy in pediatric tumor models.
- Author
-
Muldoon LL, Wu YJ, Pagel MA, and Neuwelt EA
- Subjects
- Animals, Brain Neoplasms pathology, Cells, Cultured, Child, Disease Models, Animal, Female, Humans, Male, Medulloblastoma pathology, Neoplasm Transplantation, Neuroblastoma pathology, Oxidative Stress drug effects, Rats, Rats, Nude, Acetylcysteine administration & dosage, Antineoplastic Agents toxicity, Antioxidants administration & dosage, Cisplatin toxicity, Neoplasms, Experimental drug therapy
- Abstract
Decreasing oxidative damage with the antioxidant agent N-acetylcysteine (NAC) can block the side effects of chemotherapy, but may diminish anti-tumor efficacy. We tested the potential for interactions of high dose NAC against a minimally effective cisplatin chemotherapy regimen in rat models of human pediatric cancers. Athymic rats received subcutaneous implantation of human SK-N-AS neuroblastoma cells or intra-cerebellar implantation of human D283-MED medulloblastoma cells. Rats were untreated or treated with cisplatin (3 or 4 mg/kg IV) with or without NAC (1,000 mg/kg IV) 30 min before or 4 h after cisplatin treatment. Blood urea nitrogen (BUN) and tumor volumes were measured. Cisplatin decreased the growth of SK-N-AS neuroblastoma subcutaneous tumors from 17.7 ± 4.9 to 6.4 ± 2.5 fold over baseline 2 weeks after treatment (P < 0.001). Pretreatment with NAC decreased cisplatin efficacy, while 4 h delayed NAC did not significantly affect cisplatin anti-tumor effects (relative tumor volume 6.8 ± 2.0 fold baseline, P < 0.001). In D283-MED medulloblastoma brain tumors, cisplatin decreased final tumor volume to 3.9 ± 2.3 mm(3) compared to untreated tumor volume of 45.9 ± 38.7 (P = 0.008). Delayed NAC did not significantly alter cisplatin efficacy (tumor volume 6.8 ± 8.1 mm(3), P = 0.014 versus control). Cisplatin was minimally nephrotoxic in these models. NAC decreased cisplatin-induced elevations in BUN (P < 0.02). NAC chemoprotection did not alter cisplatin therapy, if delayed until 4 h after chemotherapy. These data support a Phase I/II clinical trial of delayed NAC to reduce ototoxicity in children with localized pediatric cancers.
- Published
- 2015
- Full Text
- View/download PDF
25. Evaluation of pseudoprogression in patients with glioblastoma multiforme using dynamic magnetic resonance imaging with ferumoxytol calls RANO criteria into question.
- Author
-
Nasseri M, Gahramanov S, Netto JP, Fu R, Muldoon LL, Varallyay C, Hamilton BE, and Neuwelt EA
- Subjects
- Adult, Aged, Aged, 80 and over, Brain Neoplasms pathology, Brain Neoplasms therapy, Chemoradiotherapy methods, Combined Modality Therapy methods, Disease Progression, Female, Glioblastoma pathology, Glioblastoma therapy, Humans, Magnetic Resonance Imaging methods, Male, Middle Aged, Treatment Outcome, Young Adult, Brain Neoplasms diagnosis, Ferrosoferric Oxide therapeutic use, Glioblastoma diagnosis
- Abstract
Background: Diagnosis of pseudoprogression in patients with glioblastoma multiforme (GBM) is limited by Response Assessment in Neuro-Oncology (RANO) criteria to 3 months after chemoradiotherapy (CRT). Frequency of pseudoprogression occurring beyond this time limit was determined. Survival comparison was made between pseudoprogression and true progression patients as determined by using perfusion magnetic resonance imaging with ferumoxytol (p-MRI-Fe)., Methods: Fifty-six patients with GBM who demonstrated conventional findings concerning for progression of disease post CRT were enrolled in institutional review board-approved MRI protocols. Dynamic susceptibility-weighted contrast-enhanced p-MRI-Fe was used to distinguish true progression from pseudoprogression using relative cerebral blood volume (rCBV) values. rCBV of 1.75 was assigned as the cutoff value. Participants were followed up using RANO criteria, and survival data were analyzed., Results: Twenty-seven participants (48.2%) experienced pseudoprogression. Pseudoprogression occurred later than 3 months post CRT in 8 (29.6%) of these 27 participants (ie, 8 [14.3%] of the 56 patients meeting the inclusion criteria). Overall survival was significantly longer in participants with pseudoprogression (35.2 months) compared with those who never experienced pseudoprogression (14.3 months; P < .001)., Conclusions: Pseudoprogression presented after 3 months post CRT in a considerable portion of patients with GBM, which raises doubts about the value of the 3-month time limit of the RANO criteria. Accurate rCBV measurement (eg, p-MRI-Fe) is suggested when there are radiographical concerns about progression of disease in GBM patients, regardless of any time limit. Pseudoprogression correlates with significantly better survival outcomes., (© The Author(s) 2014. Published by Oxford University Press on behalf of the Society for Neuro-Oncology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2014
- Full Text
- View/download PDF
26. Delivery of chemotherapeutics across the blood-brain barrier: challenges and advances.
- Author
-
Doolittle ND, Muldoon LL, Culp AY, and Neuwelt EA
- Subjects
- Animals, Antineoplastic Agents therapeutic use, Central Nervous System Neoplasms drug therapy, Central Nervous System Neoplasms metabolism, Contrast Media pharmacokinetics, Humans, Lymphoma, B-Cell drug therapy, Lymphoma, B-Cell metabolism, Magnetic Resonance Imaging, Antineoplastic Agents pharmacokinetics, Blood-Brain Barrier metabolism
- Abstract
The blood-brain barrier (BBB) limits drug delivery to brain tumors. We utilize intraarterial infusion of hyperosmotic mannitol to reversibly open the BBB by shrinking endothelial cells and opening tight junctions between the cells. This approach transiently increases the delivery of chemotherapy, antibodies, and nanoparticles to brain. Our preclinical studies have optimized the BBB disruption (BBBD) technique and clinical studies have shown its safety and efficacy. The delivery of methotrexate-based chemotherapy in conjunction with BBBD provides excellent outcomes in primary central nervous system lymphoma (PCNSL) including stable or improved cognitive function in survivors a median of 12 years (range 2-26 years) after diagnosis. The addition of rituximab to chemotherapy with BBBD for PCNSL can be safely accomplished with excellent overall survival. Our translational studies of thiol agents to protect against platinum-induced toxicities led to the development of a two-compartment model in brain tumor patients. We showed that delayed high-dose sodium thiosulfate protects against carboplatin-induced hearing loss, providing the framework for large cooperative group trials of hearing chemoprotection. Neuroimaging studies have identified that ferumoxytol, an iron oxide nanoparticle blood pool agent, appears to be a superior contrast agent to accurately assess therapy-induced changes in brain tumor vasculature, in brain tumor response to therapy, and in differentiating central nervous system lesions with inflammatory components. This chapter reviews the breakthroughs, challenges, and future directions for BBBD., (© 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
27. Preservation of cognitive function in primary CNS lymphoma survivors a median of 12 years after enhanced chemotherapy delivery.
- Author
-
Doolittle ND, Dósa E, Fu R, Muldoon LL, Maron LM, Lubow MA, Tyson RM, Lacy CA, Kraemer DF, Butler RW, and Neuwelt EA
- Subjects
- Cognition Disorders chemically induced, Female, Humans, Male, Middle Aged, Neuropsychological Tests, Survivors statistics & numerical data, Antineoplastic Agents administration & dosage, Antineoplastic Agents adverse effects, Central Nervous System Neoplasms drug therapy, Cognition Disorders prevention & control, Lymphoma drug therapy
- Published
- 2013
- Full Text
- View/download PDF
28. Long-term cognitive function, neuroimaging, and quality of life in primary CNS lymphoma.
- Author
-
Doolittle ND, Korfel A, Lubow MA, Schorb E, Schlegel U, Rogowski S, Fu R, Dósa E, Illerhaus G, Kraemer DF, Muldoon LL, Calabrese P, Hedrick N, Tyson RM, Jahnke K, Maron LM, Butler RW, and Neuwelt EA
- Subjects
- Antimetabolites, Antineoplastic adverse effects, Central Nervous System Neoplasms pathology, Chemoradiotherapy, Humans, Lymphoma pathology, Methotrexate adverse effects, Neuroimaging methods, Neuropsychological Tests, Prospective Studies, Antimetabolites, Antineoplastic therapeutic use, Central Nervous System Neoplasms therapy, Cognition drug effects, Lymphoma therapy, Methotrexate therapeutic use, Quality of Life
- Abstract
Objective: To describe and correlate neurotoxicity indicators in long-term primary CNS lymphoma (PCNSL) survivors who were treated with high-dose methotrexate-based regimens with or without whole-brain radiotherapy (WBRT)., Methods: Eighty PCNSL survivors from 4 treatment groups (1 with WBRT and 3 without WBRT) who were a minimum of 2 years after diagnosis and in complete remission underwent prospective neuropsychological, quality-of-life (QOL), and brain MRI evaluation. Clinical characteristics were compared among treatments by using the χ(2) test and analysis of variance. The association among neuroimaging, neuropsychological, and QOL outcomes was assessed by using the Pearson correlation coefficient., Results: The median interval from diagnosis to evaluation was 5.5 years (minimum, 2 years; maximum, 26 years). Survivors treated with WBRT had lower mean scores in attention/executive function (p = 0.0011), motor skills (p = 0.0023), and neuropsychological composite score (p = 0.0051) compared with those treated without WBRT. Verbal memory was better in survivors with longer intervals from diagnosis to evaluation (p = 0.0045). On brain imaging, mean areas of total T2 abnormalities were different among treatments (p = 0.0006). Total T2 abnormalities after WBRT were more than twice the mean of any non-WBRT group and were associated with poorer neuropsychological and QOL outcomes., Conclusions: Our results suggest that in patients treated for PCNSL achieving complete remission and surviving at least 2 years, the addition of WBRT to methotrexate-based chemotherapy increases the risk of treatment-related neurotoxicity. Verbal memory may improve over time., Classification of Evidence: This study provides Class III evidence that in patients treated for PCNSL achieving complete remission and surviving at least 2 years, the addition of WBRT to methotrexate-based chemotherapy increases the risk of treatment-related neurotoxicity.
- Published
- 2013
- Full Text
- View/download PDF
29. Acetaminophen enhances cisplatin- and paclitaxel-mediated cytotoxicity to SKOV3 human ovarian carcinoma.
- Author
-
Wu YJ, Neuwelt AJ, Muldoon LL, and Neuwelt EA
- Subjects
- Acetaminophen therapeutic use, Animals, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Antineoplastic Combined Chemotherapy Protocols pharmacology, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Apoptosis drug effects, Cell Line, Tumor, Cisplatin therapeutic use, Female, Glutathione metabolism, Humans, Membrane Potential, Mitochondrial drug effects, NF-E2-Related Factor 2 metabolism, Neoplasm Transplantation, Paclitaxel therapeutic use, Rats, Rats, Nude, Reactive Oxygen Species, Xenograft Model Antitumor Assays, Acetaminophen pharmacology, Cisplatin pharmacology, Ovarian Neoplasms drug therapy, Paclitaxel pharmacology
- Abstract
Background: Ovarian cancer is commonly treated with cisplatin/paclitaxel but many tumors become resistant. Acetaminophen reduced glutathione and enhanced chemotherapy efficacy in hepatic cancer treatment. The objective of this study was to examine if acetaminophen enhances the cytotoxicity of cisplatin/paclitaxel in ovarian cancer., Materials and Methods: SKOV3 human ovarian carcinoma cells in vitro and a subcutaneous tumor nude rat model were used and treated with cisplatin/paclitaxel with or without acetaminophen., Results: In vitro, acetaminophen enhanced apoptosis induced by cisplatin and paclitaxel with similar effects on glutathione, reactive oxygen species and mitochondrial membrane potential, but different effects on nuclear factor erythroid 2-related factor 2 (NRF2) translocation. In vivo, acetaminophen was uniformly distributed in tissues and significantly reduced hepatic glutathione. Acetaminophen enhanced the cisplatin chemotherapeutic effect by reducing tumor recurrence., Conclusion: Our results suggest that acetaminophen as a chemoenhancing adjuvant could improve the efficacy of cisplatin and paclitaxel in treating patients with ovarian carcinoma and other tumor types.
- Published
- 2013
30. High-resolution steady-state cerebral blood volume maps in patients with central nervous system neoplasms using ferumoxytol, a superparamagnetic iron oxide nanoparticle.
- Author
-
Varallyay CG, Nesbit E, Fu R, Gahramanov S, Moloney B, Earl E, Muldoon LL, Li X, Rooney WD, and Neuwelt EA
- Subjects
- Blood Volume, Brain pathology, Brain Neoplasms blood supply, Brain Neoplasms pathology, Central Nervous System Neoplasms pathology, Cerebrovascular Circulation, Gadolinium, Heterocyclic Compounds, Humans, Organometallic Compounds, Blood Volume Determination methods, Brain blood supply, Central Nervous System Neoplasms blood supply, Contrast Media, Magnetite Nanoparticles
- Abstract
Cerebral blood volume (CBV) measurement complements conventional magnetic resonance imaging (MRI) to indicate pathologies in the central nervous system (CNS). Dynamic susceptibility contrast (DSC) perfusion imaging is limited by low resolution and distortion. Steady-state (SS) imaging may provide higher resolution CBV maps but was not previously possible in patients. We tested the feasibility of clinical SS-CBV measurement using ferumoxytol, a nanoparticle blood pool contrast agent. SS-CBV measurement was analyzed at various ferumoxytol doses and compared with DSC-CBV using gadoteridol. Ninety nine two-day MRI studies were acquired in 65 patients with CNS pathologies. The SS-CBV maps showed improved contrast to noise ratios, decreased motion artifacts at increasing ferumoxytol doses. Relative CBV (rCBV) values obtained in the thalamus and tumor regions indicated good consistency between the DSC and SS techniques when the higher dose (510 mg) ferumoxytol was used. The SS-CBV maps are feasible using ferumoxytol in a clinical dose of 510 mg, providing higher resolution images with comparable rCBV values to the DSC technique. Physiologic imaging using nanoparticles will be beneficial in visualizing CNS pathologies with high vascularity that may or may not correspond with blood-brain barrier abnormalities.
- Published
- 2013
- Full Text
- View/download PDF
31. Inhibition of SUR1 decreases the vascular permeability of cerebral metastases.
- Author
-
Thompson EM, Pishko GL, Muldoon LL, and Neuwelt EA
- Subjects
- Animals, Brain Neoplasms blood supply, Brain Neoplasms secondary, Cell Line, Tumor, Dexamethasone administration & dosage, Gene Expression, Infusions, Subcutaneous, Melanoma blood supply, Melanoma secondary, Rats, Rats, Nude, Small Cell Lung Carcinoma blood supply, Small Cell Lung Carcinoma secondary, Sulfonylurea Receptors genetics, Sulfonylurea Receptors metabolism, Tight Junctions metabolism, Xenograft Model Antitumor Assays, Zonula Occludens-1 Protein metabolism, Antimetabolites, Antineoplastic administration & dosage, Brain Neoplasms drug therapy, Capillary Permeability drug effects, Glyburide administration & dosage, Melanoma drug therapy, Small Cell Lung Carcinoma drug therapy, Sulfonylurea Receptors antagonists & inhibitors
- Abstract
Inhibition of sulfonylurea receptor 1 (SUR1) by glyburide has been shown to decrease edema after subarachnoid hemorrhage. We investigated if inhibiting SUR1 reduces cerebral edema due to metastases, the most common brain tumor, and explored the putative association of SUR1 and the endothelial tight junction protein, zona occludens-1 (ZO-1). Nude rats were intracerebrally implanted with small cell lung carcinoma (SCLC) LX1 or A2058 melanoma cells (n = 36). Rats were administered vehicle, glyburide (4.8 µg twice, orally), or dexamethasone (0.35 mg, intravenous). Blood-tumor barrier (BTB) permeability (K (trans)) was evaluated before and after treatment using dynamic contrast-enhanced magnetic resonance imaging. SUR1 and ZO-1 expression was evaluated using immunofluorescence and Western blots. In both models, SUR1 expression was significantly increased (P < .05) in tumors. In animals with SCLC, control mean K (trans) (percent change ± standard error) was 101.8 ± 36.6%, and both glyburide (-21.4 ± 14.2%, P < .01) and dexamethasone (-14.2 ± 13.1%, P < .01) decreased BTB permeability. In animals with melanoma, compared to controls (117.1 ± 43.4%), glyburide lowered BTB permeability increase (3.2 ± 15.4%, P < .05), while dexamethasone modestly lowered BTB permeability increase (63.1 ± 22.1%, P > .05). Both glyburide (P < .001) and dexamethasone (P < .01) decreased ZO-1 gap formation. By decreasing ZO-1 gaps, glyburide was at least as effective as dexamethasone at halting increased BTB permeability caused by SCLC and melanoma. Glyburide is a safe, inexpensive, and efficacious alternative to dexamethasone for the treatment of cerebral metastasis-related vasogenic edema.
- Published
- 2013
- Full Text
- View/download PDF
32. Pseudoprogression of glioblastoma after chemo- and radiation therapy: diagnosis by using dynamic susceptibility-weighted contrast-enhanced perfusion MR imaging with ferumoxytol versus gadoteridol and correlation with survival.
- Author
-
Gahramanov S, Muldoon LL, Varallyay CG, Li X, Kraemer DF, Fu R, Hamilton BE, Rooney WD, and Neuwelt EA
- Subjects
- Adult, Aged, Brain Neoplasms mortality, Chemoradiotherapy, Contrast Media, Female, Gadolinium, Humans, Male, Middle Aged, Prognosis, Reproducibility of Results, Risk Assessment, Sensitivity and Specificity, Statistics as Topic, Survival Analysis, Survival Rate, Treatment Outcome, Brain Neoplasms diagnosis, Brain Neoplasms therapy, Ferrosoferric Oxide, Heterocyclic Compounds, Magnetic Resonance Angiography methods, Organometallic Compounds
- Abstract
Purpose: To compare gadoteridol and ferumoxytol for measurement of relative cerebral blood volume (rCBV) in patients with glioblastoma multiforme (GBM) who showed progressive disease at conventional magnetic resonance (MR) imaging after chemo- and radiation therapy (hereafter, chemoradiotherapy) and to correlate rCBV with survival., Materials and Methods: Informed consent was obtained from all participants before enrollment in one of four institutional review board-approved protocols. Contrast agent leakage maps and rCBV were derived from perfusion MR imaging with gadoteridol and ferumoxytol in 19 patients with apparently progressive GBM on conventional MR images after chemoradiotherapy. Patients were classified as having high rCBV (>1.75), indicating tumor, and low rCBV (≤ 1.75), indicating pseudoprogression, for each contrast agent separately, and with or without contrast agent leakage correction for imaging with gadoteridol. Statistical analysis was performed by using Kaplan-Meier survival plots with the log-rank test and Cox proportional hazards models., Results: With ferumoxytol, rCBV was low in nine (47%) patients, with median overall survival (mOS) of 591 days, and high rCBV in 10 (53%) patients, with mOS of 163 days. A hazard ratio of 0.098 (P = .004) indicated significantly improved survival. With gadoteridol, rCBV was low in 14 (74%) patients, with mOS of 474 days, and high in five (26%), with mOS of 156 days and a nonsignificant hazard ratio of 0.339 (P = .093). Five patients with mismatched high rCBV with ferumoxytol and low rCBV with gadoteridol had an mOS of 171 days. When leakage correction was applied, rCBV with gadoteridol was significantly associated with survival (hazard ratio, 0.12; P = .003)., Conclusion: Ferumoxytol as a blood pool agent facilitates differentiation between tumor progression and pseudoprogression, appears to be a good prognostic biomarker, and unlike gadoteridol, does not require contrast agent leakage correction.
- Published
- 2013
- Full Text
- View/download PDF
33. Immunologic privilege in the central nervous system and the blood-brain barrier.
- Author
-
Muldoon LL, Alvarez JI, Begley DJ, Boado RJ, Del Zoppo GJ, Doolittle ND, Engelhardt B, Hallenbeck JM, Lonser RR, Ohlfest JR, Prat A, Scarpa M, Smeyne RJ, Drewes LR, and Neuwelt EA
- Subjects
- Animals, Endothelium, Vascular immunology, Humans, Neuroimaging, Neuroimmunomodulation, Blood-Brain Barrier immunology, Central Nervous System Diseases immunology, Neurogenic Inflammation immunology
- Abstract
The brain is in many ways an immunologically and pharmacologically privileged site. The blood-brain barrier (BBB) of the cerebrovascular endothelium and its participation in the complex structure of the neurovascular unit (NVU) restrict access of immune cells and immune mediators to the central nervous system (CNS). In pathologic conditions, very well-organized immunologic responses can develop within the CNS, raising important questions about the real nature and the intrinsic and extrinsic regulation of this immune privilege. We assess the interactions of immune cells and immune mediators with the BBB and NVU in neurologic disease, cerebrovascular disease, and intracerebral tumors. The goals of this review are to outline key scientific advances and the status of the science central to both the neuroinflammation and CNS barriers fields, and highlight the opportunities and priorities in advancing brain barriers research in the context of the larger immunology and neuroscience disciplines. This review article was developed from reports presented at the 2011 Annual Blood-Brain Barrier Consortium Meeting.
- Published
- 2013
- Full Text
- View/download PDF
34. Targeting αV-integrins decreased metastasis and increased survival in a nude rat breast cancer brain metastasis model.
- Author
-
Wu YJ, Muldoon LL, Gahramanov S, Kraemer DF, Marshall DJ, and Neuwelt EA
- Subjects
- Animals, Antibodies, Monoclonal, Humanized, Brain Neoplasms drug therapy, Brain Neoplasms metabolism, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Cell Line, Tumor, Disease Models, Animal, Female, Humans, Immunohistochemistry, Magnetic Resonance Imaging, Neoplasm Invasiveness pathology, Rats, Rats, Nude, Xenograft Model Antitumor Assays, Antibodies, Monoclonal pharmacology, Antineoplastic Agents pharmacology, Brain Neoplasms secondary, Breast Neoplasms pathology, Integrin alpha5 metabolism
- Abstract
Brain metastases commonly occur in patients with breast, lung and melanoma systemic cancers. The anti-α(V) integrin monoclonal antibody intetumumab binds cell surface proteins important for adhesion, invasion and angiogenesis in the metastatic cascade. The objective of this study was to investigate the anti-metastatic effect of intetumumab in a hematogenous breast cancer brain metastasis model. Female nude rats received intra-carotid infusion of human brain-seeking metastatic breast cancer cells (231BR-HER2) and were randomly assigned into four groups: (1) control; (2) intetumumab mixed with cells in vitro 5 min before infusion without further treatment; (3) intetumumab intravenously 4 h before and weekly after cell infusion; (4) intetumumab intravenously weekly starting 7 days after cell infusion. Brain metastases were detected by magnetic resonance imaging (MRI) and immunohistochemistry. Comparisons were made using the Kruskal-Wallis test and Dunnett's test. Survival times were estimated using Kaplan-Meier analysis. All control rats with brain tissue available for histology (9 of 11 rats) developed multiple brain metastases (median = 14). Intetumumab treatment either in vitro prior to cell infusion or intravenous before or after cell infusion prevented metastasis formation on MRI and decreased the number of metastases on histology (median = 2, p = 0.0055), including 30 % of animals without detectable tumors at the end of the study. The overall survival was improved by intetumumab compared to controls (median 77+ vs. 52 days, p = 0.0277). Our results suggest that breast cancer patients at risk of metastases might benefit from early intetumumab treatment.
- Published
- 2012
- Full Text
- View/download PDF
35. Improved perfusion MR imaging assessment of intracerebral tumor blood volume and antiangiogenic therapy efficacy in a rat model with ferumoxytol.
- Author
-
Gahramanov S, Muldoon LL, Li X, and Neuwelt EA
- Subjects
- Animals, Antibodies, Monoclonal, Humanized pharmacology, Bevacizumab, Blood Volume, Brain Neoplasms drug therapy, Brain Neoplasms pathology, Glioma drug therapy, Glioma pathology, Image Processing, Computer-Assisted, Rats, Rats, Nude, Transplantation, Heterologous, Brain Neoplasms blood supply, Cerebrovascular Circulation, Contrast Media, Ferrosoferric Oxide, Gadolinium DTPA, Glioma blood supply, Magnetic Resonance Imaging methods
- Abstract
Purpose: To evaluate the consistency of tumor blood volume measurements and antiangiogenic therapy efficacy assessments with a low-molecular-weight gadolinium-based contrast agent (GBCA, gadodiamide) versus an iron oxide nanoparticle (ferumoxytol) in the presence or absence of a loading dose of contrast agent before perfusion magnetic resonance (MR) imaging (preload method)., Materials and Methods: The protocol was approved by the institutional animal care and use committee. U87MG tumor cells were implanted intracerebrally in 13 rats. All 13 rats underwent 11.75-T MR imaging with gadodiamide (60 μL) 13 days after tumor implantation. The next day, nine rats underwent MR imaging with ferumoxytol (60 μL). Immediately after ferumoxytol imaging, six rats received bevacizumab (45 mg/kg). MR imaging was repeated 48 hours after bevacizumab treatment with gadodiamide and 72 hours after treatment with ferumoxytol. Each study included three consecutive dynamic susceptibility-weighted contrast material-enhanced (DSC) MR acquisitions, which were performed without preload, with single-dose preload, and with double-dose preload. Tumor relative cerebral blood volume (rCBV) was estimated from each DSC MR acquisition. Two-way repeated measures analysis of variance was performed to test for differences between groups with both contrast agents., Results: DSC MR imaging with gadodiamide and without preload showed low rCBV (≤ 1.75) in nine of the 13 tumors; estimated rCBV increased progressively with both single- and double-dose preloads (P < .001). Conversely, rCBVs obtained with ferumoxytol were high (>1.75) and remained constant with all three acquisitions. The magnitude of rCBV decrease after bevacizumab administration was dependent on the dose of gadodiamide preload, whereas the magnitude of rCBV decrease with ferumoxytol was constant regardless of whether contrast agent preload was used., Conclusion: With GBCA, tumor rCBV can be underestimated without preload and becomes dose dependent with preload correction. Conversely, ferumoxytol provides consistent assessment of tumor rCBV and antiangiogenic therapy efficacy., (© RSNA, 2011.)
- Published
- 2011
- Full Text
- View/download PDF
36. MRI using ferumoxytol improves the visualization of central nervous system vascular malformations.
- Author
-
Dósa E, Tuladhar S, Muldoon LL, Hamilton BE, Rooney WD, and Neuwelt EA
- Subjects
- Adult, Aged, Biomarkers, Child, Female, Ferrosoferric Oxide, Hematinics, Humans, Image Processing, Computer-Assisted, Male, Middle Aged, Central Nervous System Vascular Malformations pathology, Magnetic Resonance Imaging methods, Magnetite Nanoparticles
- Abstract
Background and Purpose: Central nervous system vascular malformations (VMs) result from abnormal vasculo- and/or angiogenesis. Cavernomas and arteriovenous malformations are also sites of active inflammation. The aim of this study was to determine whether MRI detection of VMs can be improved by administration of ferumoxytol iron oxide nanoparticle, which acts as a blood pool agent at early time points and an inflammatory marker when taken up by tissue macrophages., Methods: Nineteen patients (11 men, 8 women; mean age, 47.5 years) with central nervous system VMs underwent 3-T MRI both with gadoteridol and ferumoxytol. The ferumoxytol-induced signal changes on the T1-, T2-, and susceptibility-weighted images were analyzed at 25 minutes (range, 21 to 30 minutes) and 24 hours (range, 22 to 27 hours)., Results: Thirty-five lesions (capillary telangiectasia, n=6; cavernoma, n=21; developmental venous anomaly, n=7; arteriovenous malformation, n=1) were seen on the pre- and postgadoteridol images. The postferumoxytol susceptibility-weighted sequences revealed 5 additional VMs (3 capillary telangiectasias, 2 cavernomas) and demonstrated further tributary veins in all patients with developmental venous anomalies. The 24-hour T1 and T2 ferumoxytol-related signal abnormalities were inconsistent among patients and within VM types. No additional area of T1 or T2 enhancement was noted with ferumoxytol compared with gadoteridol in any lesion., Conclusions: Our findings indicate that the blood pool agent ferumoxytol provides important information about the number and true extent of VMs on the susceptibility-weighted MRI. The use of ferumoxytol as a macrophage imaging agent in the visualization of inflammatory cells within and around the lesions warrants further investigation.
- Published
- 2011
- Full Text
- View/download PDF
37. Imaging and therapy with rituximab anti-CD20 immunotherapy in an animal model of central nervous system lymphoma.
- Author
-
Muldoon LL, Lewin SJ, Dósa E, Kraemer DF, Pagel MA, Doolittle ND, and Neuwelt EA
- Subjects
- Animals, Antibodies, Monoclonal, Murine-Derived administration & dosage, Antibodies, Monoclonal, Murine-Derived immunology, Antigens, CD20 immunology, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Blood-Brain Barrier drug effects, Blood-Brain Barrier physiopathology, Brain blood supply, Brain drug effects, Brain pathology, Cell Line, Tumor, Central Nervous System Neoplasms pathology, Female, Humans, Lymphoma, B-Cell pathology, Magnetic Resonance Imaging, Methotrexate administration & dosage, Random Allocation, Rats, Rats, Nude, Rituximab, Survival Analysis, Treatment Outcome, Yttrium Radioisotopes, Antibodies, Monoclonal, Murine-Derived pharmacology, Central Nervous System Neoplasms drug therapy, Lymphoma, B-Cell drug therapy, Xenograft Model Antitumor Assays
- Abstract
Purpose: To evaluate the effect of rituximab monoclonal antibody (mAb) on MRI tumor volumetrics and efficacy in a rat model of central nervous system (CNS) lymphoma when delivery to the brain was optimized with osmotic blood-brain barrier disruption (BBBD)., Experimental Design: Female nude rats with intracerebral MC116 human B-cell lymphoma xenografts underwent baseline MRI and were randomized into 5 groups (n = 6 per group): (i) BBBD saline control; (ii) methotrexate with BBBD; (iii) rituximab with BBBD; (iv) rituximab and methotrexate with BBBD; and (v) intravenous rituximab. Tumor volumes were assessed by MRI at 1 week, and rats were followed for survival., Results: BBBD increased delivery of yttrium-90-radiolabeled mAb in the model of CNS lymphoma. Control rats showed 201 ± 102% increase in tumor volume on MRI 1 week after entering the study and median 14-day survival (range: 6-33). Tumor growth on MRI was slowed in the methotrexate treatment group, but survival time (median: 7 days; range: 5-12) was not different from controls. Among 17 evaluable rats treated with rituximab, 10 showed decreased tumor volume on MRI. All rituximab groups had increased survival compared with control, with a combined median of 43 days (range: 20-60, P < 0.001). There were no differences by route of delivery or combination with methotrexate., Conclusions: Rituximab was effective at decreasing tumor volume and improving survival in a model of CNS lymphoma and was not affected by combination with methotrexate or by BBBD. We suggest that rituximab warrants further study in human primary CNS lymphoma., (©2011 AACR.)
- Published
- 2011
- Full Text
- View/download PDF
38. Magnetic resonance imaging of intracranial tumors: intra-patient comparison of gadoteridol and ferumoxytol.
- Author
-
Dósa E, Guillaume DJ, Haluska M, Lacy CA, Hamilton BE, Njus JM, Rooney WD, Kraemer DF, Muldoon LL, and Neuwelt EA
- Subjects
- Adult, Aged, Blood Volume, Brain Neoplasms therapy, Female, Ferrosoferric Oxide, Gadolinium, Humans, Image Processing, Computer-Assisted, Male, Middle Aged, Prognosis, Prospective Studies, Young Adult, Brain Neoplasms diagnosis, Contrast Media, Hematinics, Heterocyclic Compounds, Magnetic Resonance Imaging, Magnetite Nanoparticles, Organometallic Compounds
- Abstract
This study aims to compare gadoteridol with ferumoxytol for contrast-enhanced and perfusion-weighted (PW) MRI of intracranial tumors. The final analysis included 26 patients, who underwent 3 consecutive days of 3T MRI. Day 1 consisted of anatomical pre- and postcontrast images, and PW MRI was acquired using gadoteridol (0.1 mmol/kg). On Day 2, the same MRI sequences were obtained with ferumoxytol (510 mg) and on Day 3, the anatomical images were repeated to detect delayed ferumoxytol-induced signal changes. The T₁-weighted images were evaluated qualitatively and quantitatively for enhancement volume and signal intensity (SI) changes; PW data were used to estimate the relative cerebral blood volume (rCBV). All 26 lesions showed 24-hour T₁-weighted ferumoxytol enhancement; 16 also had T₂-weighted hypointensities. In 6 patients, ferumoxytol-induced signal changes were noted in areas with no gadoteridol enhancement. Significantly greater (P< .0001) SI changes were seen with gadoteridol, and qualitative analyses (lesion border delineation, internal morphology, contrast enhancement) also showed significant preferences (P= .0121; P = .0015; P < .0001, respectively) for this agent. There was no significant difference in lesion enhancement volumes between contrast materials. The ferumoxytol-rCBV values were significantly higher (P = .0016) compared with the gadoteridol-rCBV values. In conclusion, ferumoxytol provides important information about tumor biology that complements gadoteridol imaging. The rCBV measurements indicate areas of tumor undergoing rapid growth, whereas the 24-hour scans mark the presence of inflammatory cells. Both of these functions provide useful information about tumor response to treatment. We suggest that dynamic and anatomical imaging with ferumoxytol warrant further assessment in brain tumor therapy.
- Published
- 2011
- Full Text
- View/download PDF
39. Potential for differentiation of pseudoprogression from true tumor progression with dynamic susceptibility-weighted contrast-enhanced magnetic resonance imaging using ferumoxytol vs. gadoteridol: a pilot study.
- Author
-
Gahramanov S, Raslan AM, Muldoon LL, Hamilton BE, Rooney WD, Varallyay CG, Njus JM, Haluska M, and Neuwelt EA
- Subjects
- Adult, Aged, Blood Volume, Blood-Brain Barrier, Brain Neoplasms blood supply, Brain Neoplasms drug therapy, Brain Neoplasms radiotherapy, Female, Ferrosoferric Oxide, Gadolinium, Glioblastoma blood supply, Glioblastoma drug therapy, Glioblastoma radiotherapy, Humans, Male, Middle Aged, Pilot Projects, Young Adult, Brain Neoplasms pathology, Contrast Media, Disease Progression, Glioblastoma pathology, Heterocyclic Compounds, Magnetic Resonance Imaging methods, Magnetite Nanoparticles, Organometallic Compounds
- Abstract
Purpose: We evaluated dynamic susceptibility-weighted contrast-enhanced magnetic resonance imaging (DSC-MRI) using gadoteridol in comparison to the iron oxide nanoparticle blood pool agent, ferumoxytol, in patients with glioblastoma multiforme (GBM) who received standard radiochemotherapy (RCT)., Methods and Materials: Fourteen patients with GBM received standard RCT and underwent 19 MRI sessions that included DSC-MRI acquisitions with gadoteridol on Day 1 and ferumoxytol on Day 2. Relative cerebral blood volume (rCBV) values were calculated from DSC data obtained from each contrast agent. T1-weighted acquisition post-gadoteridol administration was used to identify enhancing regions., Results: In seven MRI sessions of clinically presumptive active tumor, gadoteridol-DSC showed low rCBV in three and high rCBV in four, whereas ferumoxytol-DSC showed high rCBV in all seven sessions (p = 0.002). After RCT, seven MRI sessions showed increased gadoteridol contrast enhancement on T1-weighted scans coupled with low rCBV without significant differences between contrast agents (p = 0.9). Based on post-gadoteridol T1-weighted scans, DSC-MRI, and clinical presentation, four patterns of response to RCT were observed: regression, pseudoprogression, true progression, and mixed response., Conclusion: We conclude that DSC-MRI with a blood pool agent such as ferumoxytol may provide a better monitor of tumor rCBV than DSC-MRI with gadoteridol. Lesions demonstrating increased enhancement on T1-weighted MRI coupled with low ferumoxytol rCBV are likely exhibiting pseudoprogression, whereas high rCBV with ferumoxytol is a better marker than gadoteridol for determining active tumor. These interesting pilot observations suggest that ferumoxytol may differentiate tumor progression from pseudoprogression and warrant further investigation., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
40. Dynamic magnetic resonance imaging assessment of vascular targeting agent effects in rat intracerebral tumor models.
- Author
-
Muldoon LL, Gahramanov S, Li X, Marshall DJ, Kraemer DF, and Neuwelt EA
- Subjects
- Angiogenesis Inhibitors therapeutic use, Animals, Antibodies, Monoclonal, Humanized, Bevacizumab, Brain Neoplasms drug therapy, Capillary Permeability, Female, Gadolinium DTPA, Humans, Integrin alphaV chemistry, Integrin alphaV immunology, Rats, Rats, Nude, Tumor Cells, Cultured, Vascular Endothelial Growth Factor A antagonists & inhibitors, Vascular Endothelial Growth Factor A immunology, Antibodies, Monoclonal therapeutic use, Brain Neoplasms blood supply, Brain Neoplasms pathology, Disease Models, Animal, Magnetic Resonance Imaging, Neovascularization, Pathologic prevention & control
- Abstract
We used dynamic MRI to evaluate the effects of monoclonal antibodies targeting brain tumor vasculature. Female athymic rats with intracerebral human tumor xenografts were untreated or treated with intetumumab, targeting α(V)-integrins, or bevacizumab, targeting vascular endothelial growth factor (n = 4-6 per group). Prior to treatment and at 1, 3, and 7 days after treatment, we performed standard MRI to assess tumor volume, dynamic susceptibility-contrast MRI with the blood-pool iron oxide nanoparticle ferumoxytol to evaluate relative cerebral blood volume (rCBV), and dynamic contrast-enhanced MRI to assess tumor vascular permeability. Tumor rCBV increased by 27 ± 13% over 7 days in untreated rats; intetumumab increased tumor rCBV by 65 ± 10%, whereas bevacizumab reduced tumor rCBV by 31 ± 10% at 7 days (P < .001 for group and day). Similarly, intetumumab increased brain tumor vascular permeability compared with controls at 3 and 7 days after treatment, whereas bevacizumab decreased tumor permeability within 24 hours (P = .0004 for group, P = .0081 for day). All tumors grew over the 7-day assessment period, but bevacizumab slowed the increase in tumor volume on MRI. We conclude that the vascular targeting agents intetumumab and bevacizumab had diametrically opposite effects on dynamic MRI of tumor vasculature in rat brain tumor models. Targeting α(V)-integrins increased tumor vascular permeability and blood volume, whereas bevacizumab decreased both measures. These findings have implications for chemotherapy delivery and antitumor efficacy.
- Published
- 2011
- Full Text
- View/download PDF
41. Dynamic-contrast-enhanced-MRI with extravasating contrast reagent: rat cerebral glioma blood volume determination.
- Author
-
Li X, Rooney WD, Várallyay CG, Gahramanov S, Muldoon LL, Goodman JA, Tagge IJ, Selzer AH, Pike MM, Neuwelt EA, and Springer CS Jr
- Subjects
- Animals, Blood Volume, Cell Line, Tumor, Computer Simulation, Contrast Media pharmacokinetics, Glioma blood supply, Image Interpretation, Computer-Assisted methods, Magnetic Resonance Imaging, Male, Metabolic Clearance Rate, Models, Neurological, Rats, Rats, Nude, Blood Volume Determination methods, Brain Neoplasms metabolism, Brain Neoplasms pathology, Gadolinium DTPA pharmacokinetics, Glioma metabolism, Glioma pathology
- Abstract
The accurate mapping of the tumor blood volume (TBV) fraction (vb) is a highly desired imaging biometric goal. It is commonly thought that achieving this is difficult, if not impossible, when small molecule contrast reagents (CRs) are used for the T1-weighted (Dynamic-Contrast-Enhanced) DCE-MRI technique. This is because angiogenic malignant tumor vessels allow facile CR extravasation. Here, a three-site equilibrium water exchange model is applied to DCE-MRI data from the cerebrally-implanted rat brain U87 glioma, a tumor exhibiting rapid CR extravasation. Analyses of segments of the (and the entire) DCE data time-course with this "shutter-speed" pharmacokinetic model, which admits finite water exchange kinetics, allow TBV estimation from the first-pass segment. Pairwise parameter determinances were tested with grid searches of 2D parametric error surfaces. Tumor blood volume (vb), as well as ve (the extracellular, extravascular space volume fraction), and Ktrans (a CR extravasation rate measure) parametric maps are presented. The role of the Patlak Plot in DCE-MRI is also considered., (Copyright © 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
42. Superparamagnetic iron oxide nanoparticles: diagnostic magnetic resonance imaging and potential therapeutic applications in neurooncology and central nervous system inflammatory pathologies, a review.
- Author
-
Weinstein JS, Varallyay CG, Dosa E, Gahramanov S, Hamilton B, Rooney WD, Muldoon LL, and Neuwelt EA
- Subjects
- Animals, Blood-Brain Barrier, Contrast Media, Electron Spin Resonance Spectroscopy, Humans, Nanoparticles, Particle Size, Central Nervous System Neoplasms diagnosis, Central Nervous System Neoplasms pathology, Ferric Compounds pharmacology, Ferric Compounds therapeutic use, Inflammation diagnosis, Inflammation pathology, Magnetic Resonance Imaging methods
- Abstract
Superparamagnetic iron oxide nanoparticles have diverse diagnostic and potential therapeutic applications in the central nervous system (CNS). They are useful as magnetic resonance imaging (MRI) contrast agents to evaluate: areas of blood-brain barrier (BBB) dysfunction related to tumors and other neuroinflammatory pathologies, the cerebrovasculature using perfusion-weighted MRI sequences, and in vivo cellular tracking in CNS disease or injury. Novel, targeted, nanoparticle synthesis strategies will allow for a rapidly expanding range of applications in patients with brain tumors, cerebral ischemia or stroke, carotid atherosclerosis, multiple sclerosis, traumatic brain injury, and epilepsy. These strategies may ultimately improve disease detection, therapeutic monitoring, and treatment efficacy especially in the context of antiangiogenic chemotherapy and antiinflammatory medications. The purpose of this review is to outline the current status of superparamagnetic iron oxide nanoparticles in the context of biomedical nanotechnology as they apply to diagnostic MRI and potential therapeutic applications in neurooncology and other CNS inflammatory conditions.
- Published
- 2010
- Full Text
- View/download PDF
43. Efficacy and MRI of rituximab and methotrexate treatment in a nude rat model of CNS lymphoma.
- Author
-
Jahnke K, Muldoon LL, Varallyay CG, Lewin SJ, Brown RD, Kraemer DF, Soussain C, and Neuwelt EA
- Subjects
- Animals, Antibodies, Monoclonal, Murine-Derived, Cell Line, Tumor, Female, Humans, Image Enhancement, Rats, Rats, Nude, Rituximab, Treatment Outcome, Xenograft Model Antitumor Assays, Antibodies, Monoclonal administration & dosage, Antineoplastic Agents administration & dosage, Central Nervous System Neoplasms drug therapy, Lymphoma, B-Cell drug therapy, Magnetic Resonance Imaging methods, Methotrexate administration & dosage
- Abstract
To determine the efficacy of methotrexate and/or rituximab in a CNS lymphoma model and to evaluate MRI modalities for monitoring efficacy, we inoculated female athymic nude rats (rnu/rnu) intracerebrally with human MC116 B-lymphoma cells. Between days 16 and 26, rats were randomized to receive intravenous (IV) treatment with (1) saline (controls, n = 15), (2) methotrexate 1,000 mg/m(2) (n = 6), (3) rituximab 375 mg/m(2) (n = 6), or (4) rituximab plus methotrexate (n = 6). T2/fluid-attenuated inversion recovery (FLAIR) and gadolinium contrast-enhanced T1 MRI sequences were performed prior to and 1 week after treatment. IV rituximab gave an objective tumor response in four of six animals (>50% reduction in tumor volume comparing pre- and posttreatment T2/FLAIR MRI) and resulted in stable disease (50%-125% of baseline) in another animal. The percent change in tumor volume on T2/FLAIR images was significantly different in the control versus rituximab group (p = 0.0051). IV methotrexate slowed tumor growth, compared to controls, but only one of six animals had an objective response. In untreated controls, tumor histological volumes correlated well with T2/FLAIR or contrast-enhanced T1 images (r = 0.877). In the treatment groups, T2/FLAIR correlation was good, but the gadolinium-enhanced T1 MRI was not significantly correlated with histology (r = 0.19). The MC116 CNS lymphoma model seems valuable for preclinical testing of efficacy and toxicity of treatment regimens. IV rituximab was highly effective, but methotrexate was only minimally effective. T2/FLAIR was superior to contrast-enhanced T1 for monitoring efficacy.
- Published
- 2009
- Full Text
- View/download PDF
44. Dynamic MRI using iron oxide nanoparticles to assess early vascular effects of antiangiogenic versus corticosteroid treatment in a glioma model.
- Author
-
Varallyay CG, Muldoon LL, Gahramanov S, Wu YJ, Goodman JA, Li X, Pike MM, and Neuwelt EA
- Subjects
- Animals, Antibodies, Monoclonal therapeutic use, Antibodies, Monoclonal, Humanized, Bevacizumab, Capillary Permeability, Cerebrovascular Circulation drug effects, Dexamethasone therapeutic use, Disease Models, Animal, Drug Monitoring methods, Gadolinium DTPA, Glioma diagnosis, Humans, Kinetics, Rats, Adrenal Cortex Hormones therapeutic use, Angiogenesis Inhibitors therapeutic use, Ferric Compounds, Glioma drug therapy, Magnetic Resonance Imaging methods, Nanoparticles
- Abstract
The vascular effects of antiangiogenic treatment may pose problems for evaluating brain tumor response based on contrast-enhanced magnetic resonance imaging (MRI). We used serial dynamic contrast-enhanced MRI at 12 T to assess vascular responses to antiangiogenic versus steroid therapy. Athymic rats with intracerebral U87MG human glioma (n=17) underwent susceptibility-weighted perfusion MRI with ferumoxytol, a solely intravascular ultrasmall superparamagnetic iron oxide (USPIO) nanoparticle, followed by T1-weighted dynamic gadodiamide-enhanced MRI to measure vascular permeability. Rats were imaged before and after 24, 48, and 72 h of treatment with the antiangiogenic agent bevacizumab or the corticosteroid dexamethasone. Contrast agent extravasation was seen rapidly after gadodiamide, but not with ferumoxytol administration. Bevacizumab significantly decreased the blood volume and decreased permeability in tumors as determined by increased time-to-peak enhancement. A single dose of 45 mg/kg bevacizumab resulted in changes analogous to dexamethasone given in an extremely high dose (12 mg/kg per day), and was significantly more effective than dexamethasone at 2 mg/kg per day. We conclude that dynamic perfusion MRI measurements with ferumoxytol USPIO to assess cerebral blood volume, along with dynamic gadodiamide-enhanced MR to assess vascular permeability, hold promise in more accurately detecting therapeutic responses to antiangiogenic therapy.
- Published
- 2009
- Full Text
- View/download PDF
45. Bevacizumab and carboplatin increase survival and asymptomatic tumor volume in a glioma model.
- Author
-
Jahnke K, Muldoon LL, Varallyay CG, Lewin SJ, Kraemer DF, and Neuwelt EA
- Subjects
- Animals, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal, Humanized, Bevacizumab, Brain Neoplasms mortality, Brain Neoplasms pathology, Carboplatin administration & dosage, Glioma mortality, Glioma pathology, Humans, Magnetic Resonance Imaging, Male, Rats, Rats, Nude, Survival Rate, Tumor Burden, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Brain Neoplasms drug therapy, Disease Models, Animal, Glioma drug therapy
- Abstract
To evaluate efficacy and MRI findings after intravenous bevacizumab and/or carboplatin in a human glioma animal model, we randomized male nude rats with intracerebral UW28 human glioma xenografts to four groups: (1) controls (n = 9), (2) bevacizumab 10 mg/kg (n = 6), (3) carboplatin 200 mg/m(2) (n = 6), and (4) bevacizumab + carboplatin (n = 6). MRI was performed on the day of treatment (day 7-10) and 1 week later, and rats were followed for survival. Dynamic MRI was done in three controls and three rats treated with bevacizumab with or without carboplatin before and 24 h after treatment. Median overall survival (OS) was as follows: group 1, 16 days; group 2, 23 days; group 3, 22 days; group 4, 36 days. OS was significantly longer in group 4 than in group 1 (p = 0.0011), group 2 (p = 0.0014), and group 3 (p = 0.0015), and rats had significantly larger tumors. No objective tumor responses were observed on MR images at 1 week after treatment; however, after bevacizumab, dynamic MRI showed reduced gadolinium enhancement intensity and increased time to peak, consistent with decreased vascular permeability. Carboplatin + bevacizumab is effective and superior over bevacizumab or carboplatin monotherapy in this animal model. Increased survival concomitant with increased asymptomatic tumor volume is suggestive that vascular targeting with reduced peritumoral edema and mass effect contributes to the efficacy of bevacizumab. The promising survival data warrant future clinical trials using bevacizumab + carboplatin.
- Published
- 2009
- Full Text
- View/download PDF
46. Cyclophosphamide enhances human tumor growth in nude rat xenografted tumor models.
- Author
-
Wu YJ, Muldoon LL, Dickey DT, Lewin SJ, Varallyay CG, and Neuwelt EA
- Subjects
- Animals, Brain Neoplasms drug therapy, Brain Neoplasms secondary, Cell Proliferation drug effects, Glioma drug therapy, Humans, Magnetic Resonance Imaging, Rats, Rats, Nude, Brain Neoplasms pathology, Cyclophosphamide pharmacology, Disease Models, Animal, Glioma pathology, Xenograft Model Antitumor Assays
- Abstract
The effect of the immunomodulatory chemotherapeutic agent cyclophosphamide (CTX) on tumor growth was investigated in primary and metastatic intracerebral and subcutaneous rat xenograft models. Nude rats were treated with CTX (100 mg/kg, intraperitoneally) 24 hours before human ovarian carcinoma (SKOV3), small cell lung carcinoma (LX-1 SCLC), and glioma (UW28, U87MG, and U251) tumor cells were inoculated subcutaneously, intraperitoneally, or in the right cerebral hemisphere or were infused into the right internal carotid artery. Tumor development was monitored and recorded. Potential mechanisms were further investigated. Only animals that received both CTX and Matrigel showed consistent growth of subcutaneous tumors. Cyclophosphamide pretreatment increased the percentage (83.3% vs 0%) of animals showing intraperitoneal tumors. In intracerebral implantation tumor models, CTX pretreatment increased the tumor volume and the percentage of animals showing tumors. Cyclophosphamide increased lung carcinoma bone and facial metastases after intra-arterial injection, and 20% of animals showed brain metastases. Cyclophosphamide transiently decreased nude rat white blood cell counts and glutathione concentration, whereas serum vascular endothelial growth factor was significantly elevated. Cyclophosphamide also increased CD31 reactivity, a marker of vascular endothelium, and macrophage (CD68-positive) infiltration into glioma cell-inoculated rat brains. Cyclophosphamide may enhance primary and metastatic tumor growth through multiple mechanisms, including immune modulation, decreased response to oxidative stress, increased tumor vascularization, and increased macrophage infiltration. These findings may be clinically relevant because chemotherapy may predispose human cancer subjects to tumor growth in the brain or other tissues.
- Published
- 2009
- Full Text
- View/download PDF
47. Effect of N-acetylcysteine route of administration on chemoprotection against cisplatin-induced toxicity in rat models.
- Author
-
Dickey DT, Muldoon LL, Doolittle ND, Peterson DR, Kraemer DF, and Neuwelt EA
- Subjects
- Administration, Oral, Animals, Dose-Response Relationship, Drug, Drug Administration Schedule, Female, Injections, Intra-Arterial, Injections, Intraperitoneal, Injections, Intravenous, Kidney Diseases chemically induced, Kidney Function Tests, Rats, Rats, Long-Evans, Acetylcysteine administration & dosage, Acetylcysteine pharmacology, Acetylcysteine therapeutic use, Antineoplastic Agents toxicity, Cisplatin toxicity, Free Radical Scavengers administration & dosage, Free Radical Scavengers pharmacology, Free Radical Scavengers therapeutic use, Kidney Diseases prevention & control
- Abstract
Dosing and route of administration of N-acetylcysteine (NAC) for protection against cisplatin (CDDP) nephrotoxicity was investigated in rats. Two models of toxicity were tested: a single high dose of CDDP (10 mg/kg intraperitoneally (IP)), and multiple low dose treatments (1 mg/kg IP twice a day for 4 days, 10 days rest, then repeated). NAC (50-1,200 mg/kg) was given to the rats by IP, oral (PO), intravenous (IV) and intra-arterial (IA) routes. Renal toxicity was determined by blood urea nitrogen (BUN) and creatinine (CR) levels 3 days after treatment. Blood collected 15 min after NAC was analyzed for total NAC. Both models of CDDP administration produced renal toxicity. In the single dose CDDP model, NAC 400 mg/kg given IP and PO produced no renal protection as measured by BUN (131.8 +/- 8.2 and 123.3 +/- 8.2, respectively) or CR (2.3 +/- 0.38 and 1.77 +/- 0.21, respectively). IV NAC reduced nephrotoxicity, (BUN 26.3 +/- 6.8, CR 0.47 +/- 0.15). NAC 50 mg/kg IA gave better protection than IV. In the repeated-dose CDDP model, nephrotoxicity was blocked by 800 mg/kg NAC given IV but not IP. Blood concentrations of total NAC showed a dose response after IV NAC, but high dose NAC (1,200 mg/kg) by the PO route gave very low levels of NAC. Thus the protective properties of NAC are affected by the dose and route of administration.
- Published
- 2008
- Full Text
- View/download PDF
48. N-acetylcysteine use to prevent contrast medium-induced nephropathy: premature phase III trials.
- Author
-
Stenstrom DA, Muldoon LL, Armijo-Medina H, Watnick S, Doolittle ND, Kaufman JA, Peterson DR, Bubalo J, and Neuwelt EA
- Subjects
- Acetylcysteine pharmacology, Acetylcysteine toxicity, Clinical Trials, Phase III as Topic, Humans, Acetylcysteine therapeutic use, Contrast Media adverse effects, Kidney Diseases chemically induced, Kidney Diseases prevention & control
- Abstract
To date there has been no general consensus regarding the effectiveness of N-acetylcysteine as a protective therapy against contrast medium-induced nephropathy. Several phase III clinical trials have been conducted without a proper understanding of N-acetylcysteine pharmacology, particularly with regard to first-pass hepatic metabolism. A review was conducted of the literature concerning contrast medium-induced nephropathy and new studies of human N-acetylcysteine pharmacology were performed. After an analysis was performed, it was concluded that further phase I and phase II trials are needed. The efficacy of N-acetylcysteine in the prevention of contrast medium-induced nephropathy may be demonstrated with the use of higher doses than used in earlier studies, in combination with parenteral administration.
- Published
- 2008
- Full Text
- View/download PDF
49. Intraarterial chemotherapy and osmotic blood-brain barrier disruption for patients with embryonal and germ cell tumors of the central nervous system.
- Author
-
Jahnke K, Kraemer DF, Knight KR, Fortin D, Bell S, Doolittle ND, Muldoon LL, and Neuwelt EA
- Subjects
- Adolescent, Adult, Antineoplastic Combined Chemotherapy Protocols adverse effects, Carboplatin administration & dosage, Carboplatin adverse effects, Central Nervous System Neoplasms radiotherapy, Child, Child, Preschool, Combined Modality Therapy, Cyclophosphamide administration & dosage, Cyclophosphamide adverse effects, Disease Progression, Etoposide administration & dosage, Etoposide adverse effects, Female, Humans, Infant, Infusions, Intra-Arterial methods, Male, Methotrexate administration & dosage, Methotrexate adverse effects, Middle Aged, Neoplasms, Germ Cell and Embryonal radiotherapy, Prospective Studies, Survival Analysis, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Blood-Brain Barrier physiopathology, Central Nervous System Neoplasms drug therapy, Neoplasms, Germ Cell and Embryonal drug therapy
- Abstract
Background: The rate of durable responses in embryonal and certain germ cell tumors of the central nervous system (CNS) is unsatisfactory. Intraarterial chemotherapy and osmotic blood-brain barrier disruption (IA/BBBD) increases drug delivery to the CNS., Methods: Data of patients treated with carboplatin or methotrexate-based IA/BBBD on prospective phase 2 trials conducted at 3 centers were collected. Study outcomes included overall survival (OS), time to progression (TTP), and toxicity., Results: Fifty-four patients were treated. Twenty-seven patients received IA/BBBD as salvage treatment. The median OS was 2.8 years for all patients, 2.5 years for supratentorial and disseminated primitive neuroectodermal tumors (PNETs, n = 29), 1.7 years for medulloblastomas (n = 12), and 5.4 years for germ cell tumors (n = 13). OS and TTP for all patients were better with a Karnofsky Performance Status > or =70% (P = .0013 and .0070) and IA/BBBD as first-line treatment (P = .0059 and .029). In PNETs, OS was higher with pineal location (P = .045) and IA/BBBD as first-line treatment (P = .0036), and TTP was improved with radiotherapy before IA/BBBD (P = .036) and IA/BBBD as first-line treatment (P = .0079). Seventeen of 54 patients (31%) are alive, and 16 are alive at 4+ to 18+ years. Three survivors were not treated with radiotherapy and 4 were treated with focal radiotherapy only. The patients who were not irradiated did not develop dementia., Conclusions: Survival and toxicity data appear promising, considering the cohort's adverse prognostic profile. A plateau in survival curves suggests a cure for some patients. Long-term survival may be achieved with focal or reduced-dose radiotherapy in some IA/BBBD patients.
- Published
- 2008
- Full Text
- View/download PDF
50. In vivo leukocyte labeling with intravenous ferumoxides/protamine sulfate complex and in vitro characterization for cellular magnetic resonance imaging.
- Author
-
Wu YJ, Muldoon LL, Varallyay C, Markwardt S, Jones RE, and Neuwelt EA
- Subjects
- Animals, Antigens, CD analysis, Brain cytology, Brain metabolism, Brain surgery, Cell Movement, Contrast Media administration & dosage, Contrast Media toxicity, Dextrans, Female, Ferrosoferric Oxide administration & dosage, Ferrosoferric Oxide toxicity, Flow Cytometry, Immunophenotyping, Injections, Intravenous, Iron administration & dosage, Iron toxicity, Leukocytes, Mononuclear drug effects, Leukocytes, Mononuclear immunology, Leukocytes, Mononuclear transplantation, Magnetite Nanoparticles, Oxides administration & dosage, Oxides toxicity, Protamines administration & dosage, Protamines toxicity, Rats, Rats, Long-Evans, Time Factors, Tissue Distribution, Contrast Media metabolism, Ferrosoferric Oxide metabolism, Iron metabolism, Leukocytes, Mononuclear metabolism, Magnetic Resonance Imaging methods, Oxides metabolism, Protamines metabolism, Staining and Labeling methods
- Abstract
Cellular labeling with ferumoxides (Feridex IV) superparamagnetic iron oxide nanoparticles can be used to monitor cells in vivo by MRI. The objective of this study was to use histology and MRI to evaluate an in vivo, as opposed to in vitro, technique for labeling of mononuclear leukocytes as a means of tracking inflammatory processes in the brain. Long-Evans rats were intravenously injected with 20 mg/kg ferumoxides, ferumoxtran-10, or ferumoxytol with or without protamine sulfate. Leukocytes and splenocytes were evaluated by cell sorting and iron histochemistry or were implanted into the brain for MRI. Injection of ferumoxides/protamine sulfate complex IV resulted in iron labeling of leukocytes (ranging from 7.4 +/- 0.5% to 12.5 +/- 0.9% with average 9.2 +/- 0.8%) compared with ferumoxides (ranging from 3.9 +/- 0.4% to 6.3 +/- 0.5% with average 5.0 +/- 0.5%) or protamine sulfate alone (ranging from 0% to 0.9 +/- 0.7% with average 0.3 +/- 0.3%). Cell sorting analysis indicated that iron-labeled cells were enriched for cell types positive for the myelomonocytic marker (CD11b/c) and the B lymphocyte marker (CD45RA) and depleted in the T cell marker (CD3). Neither ferumoxtran-10 nor ferumoxytol with protamine sulfate labeled leukocytes. In vivo ferumoxides/protamine sulfate-loaded leukocytes and splenocytes were detected by MRI after intracerebral injection. Ferumoxides/protamine complex labeled CD45RA-positive and CD11b/c-positive leukocytes in vivo without immediate toxicity. The dose of feumoxides in this report is much higher than the approved human dose, so additional animal studies are required before this approach could be translated to the clinic. These results might provide useful information for monitoring leukocyte trafficking into the brain.
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.