39 results on '"Frances Chow"'
Search Results
2. Multi-omic profiling reveals discrepant immunogenic properties and a unique tumor microenvironment among melanoma brain metastases
- Author
-
Gino K. In, Jennifer R. Ribeiro, Jun Yin, Joanne Xiu, Matias A. Bustos, Fumito Ito, Frances Chow, Gabriel Zada, Lindsay Hwang, April K. S. Salama, Soo J. Park, Justin C. Moser, Sourat Darabi, Evidio Domingo-Musibay, Maria L. Ascierto, Kim Margolin, Jose Lutzky, Geoffrey T. Gibney, Michael B. Atkins, Benjamin Izar, Dave S. B. Hoon, and Ari M. VanderWalde
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Melanoma brain metastases (MBM) are clinically challenging to treat and exhibit variable responses to immune checkpoint therapies. Prior research suggests that MBM exhibit poor tumor immune responses and are enriched in oxidative phosphorylation. Here, we report results from a multi-omic analysis of a large, real-world melanoma cohort. MBM exhibited lower interferon-gamma (IFNγ) scores and T cell-inflamed scores compared to primary cutaneous melanoma (PCM) or extracranial metastases (ECM), which was independent of tumor mutational burden. Among MBM, there were fewer computationally inferred immune cell infiltrates, which correlated with lower TNF and IL12B mRNA levels. Ingenuity pathway analysis (IPA) revealed suppression of inflammatory responses and dendritic cell maturation pathways. MBM also demonstrated a higher frequency of pathogenic PTEN mutations and angiogenic signaling. Oxidative phosphorylation (OXPHOS) was enriched in MBM and negatively correlated with NK cell and B cell-associated transcriptomic signatures. Modulating metabolic or angiogenic pathways in MBM may improve responses to immunotherapy in this difficult-to-treat patient subset.
- Published
- 2023
- Full Text
- View/download PDF
3. Neoadjuvant PD-1 blockade induces T cell and cDC1 activation but fails to overcome the immunosuppressive tumor associated macrophages in recurrent glioblastoma
- Author
-
Alexander H. Lee, Lu Sun, Aaron Y. Mochizuki, Jeremy G. Reynoso, Joey Orpilla, Frances Chow, Jenny C. Kienzler, Richard G. Everson, David A. Nathanson, Steven J. Bensinger, Linda M. Liau, Timothy Cloughesy, Willy Hugo, and Robert M. Prins
- Subjects
Science - Abstract
Immune-checkpoint blockade has shown limited benefits in patients with glioblastoma. To understand how the composition of the tumor immune microenvironment might limit clinical responses, here the authors present a high dimensional profiling of the immune landscape in patients with glioblastoma following neoadjuvant PD-1 checkpoint blockade.
- Published
- 2021
- Full Text
- View/download PDF
4. 421 A Phase 1 and Randomized Phase 2 Clinical Trial of Selinexor and Temozolomide in Recurrent Glioblastoma Among Adults: The Product of a Successful Team Science Approach
- Author
-
Frances Chow, Michael Berens, Sharon Tamir, Yosef Landesman, Christopher Walker, Mark Krailo, Steven Gore, and Jana Portnow
- Subjects
Medicine - Abstract
OBJECTIVES/GOALS: Selinexor is a novel XPO1 inhibitor that blocks nuclear export, thus impairing DNA repair and causing apoptosis. Our goal was to conduct preclinical and clinical studies to test our hypothesis that selinexor’s efficacy is boosted by priming with temozolomide and is associated with a tissue biomarker. METHODS/STUDY POPULATION: We leveraged a team science approach through the NCI Cancer Therapy Evaluation Program (CTEP) to design preclinical experiments, develop a novel RNAseq analysis pipeline, and use pre-existing clinical experience to open an early phase clinical trial for recurrent glioblastoma. Team members included a CTEP medical officer, cancer biologist, pharmacist, industry scientist, translational scientist, and early career clinician scientist mentored by an expert clinician scientist. Based on preclinical results, participants in the clinical trial experimental arm will receive sequential temozolomide 150mg/m2 on days 1-5 and a starting dose of selinexor 60mg on days 8 and 15 of a 28-day cycle. Participants in the control arm will receive monotherapy temozolomide. RESULTS/ANTICIPATED RESULTS: Sequential treatment of U87 cells and intracranial xenografts had superior DNA damage (É£H2A.X, cleaved PARP) and overall survival compared to combination or single-agent (HR 0.25 [95% CI, 0.07-0.84]; p=0.01, log-rank). We used the top-scoring gene pair method to identify an RNAseq signature associated with response to selinexor. We then designed a trial for first recurrent MGMT methylated glioblastoma. Primary objectives are safety and preliminary efficacy. Secondary objectives are overall response rate, efficacy, and validation of a molecular signature. Phase 1 dose finding (n=12) will be followed by a randomized phase 2 (n=72); using proportional hazards regression, RHR 0.5 with p DISCUSSION/SIGNIFICANCE: The NCI CTEP Project Team employs team science as a framework to successfully develop multidisciplinary collaborations, build investigator trial experience, and lead the way to future research opportunities. Our trial addresses a significant unmet need to offer novel therapies and molecular biomarkers in glioblastoma.
- Published
- 2023
- Full Text
- View/download PDF
5. The abscopal effect: systematic review in patients with brain and spine metastases
- Author
-
Dhiraj J Pangal, Benjamin Yarovinsky, Tyler Cardinal, David J Cote, Jacob Ruzevick, Frank J Attenello, Eric L Chang, Jason Ye, Josh Neman, Frances Chow, and Gabriel Zada
- Subjects
General Medicine - Abstract
Background The abscopal effect is a rare phenomenon whereby local radiation induces a proposed immune-mediated anti-tumor effect at distant sites. Given the growing use of immunotherapies and systemic immune checkpoint inhibitors in neuro-oncologic practice, we aimed to review prior studies pertaining to this phenomenon in the context of tumor shrinkage both within the central nervous system as well as distant disease sites. Methods A systematic review in accordance with the PRISMA guidelines was conducted to identify all studies which assessed the abscopal effect in patients with treated metastatic cancer to the brain and/or spine. Articles were included if they reported the abscopal effect in patients (case studies) or if the abscopal effect was explicitly analyzed in case series with cohorts of patients with metastatic brain or spine tumors. Laboratory investigations and clinical trials investigating new therapies were excluded. Results Twenty reports met inclusion criteria [16 case reports, 4 case series (n = 160), total n = 174]. Case reports of the abscopal effect were in relation to the following cancers: melanoma (6 patients), breast cancer (3), lung adenocarcinoma (2), non-small-cell lung cancer (2), hepatocellular carcinoma (1), and renal cell carcinoma (1). Eleven patients had irradiation to the brain and 2 to the spine. Patients undergoing whole brain radiotherapy (6) had an average dose of 33.6 Gy over 8–15 fractions, and those undergoing stereotactic radiosurgery (5) had an average dose of 21.5 Gy over 1–5 fractions. One patient had radiation to the body and an intracranial abscopal effect was observed. Most common sites of extracranial tumor reduction were lung and lymph nodes. Ten case studies (57%) showed complete resolution of extra-CNS tumor burden. Median progression-free survival was 13 months following radiation. Four papers investigated incidence of abscopal effects in patients with metastatic melanoma to the brain who received immune checkpoint inhibitor therapy (n = 160); two papers found an abscopal effect in 35% and 52% of patients (n = 16, 21 respectively), and two papers found no evidence of abscopal effects (n = 61, 62). Conclusions Abscopal effects can occur following radiotherapy in patients with brain or spine metastases and is thought to be a result of increased anti-tumor immunity. The potential for immune checkpoint inhibitor therapy to be used in combination with radiotherapy to induce an abscopal effect is an area of active investigation.
- Published
- 2022
6. Modulating glioblastoma chemotherapy response: Evaluating long non-coding RNA effects on DNA damage response, glioma stem cell function, and hypoxic processes
- Author
-
Edith Yuan, Kristie Liu, Justin Lee, Kathleen Tsung, Frances Chow, and Frank J Attenello
- Subjects
Oncology ,Surgery ,Neurology (clinical) - Abstract
Glioblastoma (GBM) is the most common and aggressive primary adult brain tumor, with an estimated annual incidence of 17 000 new cases in the United States. Current treatments for GBM include chemotherapy, surgical resection, radiation therapy, and antiangiogenic therapy. However, despite the various therapeutic options, the 5-year survival rate remains at a dismal 5%. Temozolomide (TMZ) is the first-line chemotherapy drug for GBM; however, poor TMZ response is one of the main contributors to the dismal prognosis. Long non-coding RNAs (lncRNAs) are nonprotein coding transcripts greater than 200 nucleotides that have been implicated to mediate various GBM pathologies, including chemoresistance. In this review, we aim to frame the TMZ response in GBM via exploration of the lncRNAs mediating three major mechanisms of TMZ resistance: (1) regulation of the DNA damage response, (2) maintenance of glioma stem cell identity, and (3) exploitation of hypoxia-associated responses.
- Published
- 2022
7. Cancer stem cell assay-guided chemotherapy improves survival of patients with recurrent glioblastoma in a randomized trial
- Author
-
Tulika Ranjan, Soma Sengupta, Michael J. Glantz, Richard M. Green, Alexander Yu, Dawit Aregawi, Rekha Chaudhary, Ricky Chen, Mario Zuccarello, Christine Lu-Emerson, Hugh D. Moulding, Neil Belman, Jon Glass, Aaron Mammoser, Mark Anderson, Jagan Valluri, Nicholas Marko, Jason Schroeder, Steven Jubelirer, Frances Chow, Pier Paolo Claudio, Anthony M. Alberico, Seth T. Lirette, Krista L. Denning, and Candace M. Howard
- Subjects
General Biochemistry, Genetics and Molecular Biology - Published
- 2023
- Full Text
- View/download PDF
8. CNSC-02. BREAST TO BRAIN METASTASIS IS EXACERBATED WITH CHEMOTHERAPY THROUGH BLOOD-CEREBRAL SPINAL FLUID-BARRIER AND INDUCES ALZHEIMER’S-LIKE PATHOLOGY
- Author
-
Josh Neman, Behnaz Saatian, Krutika Deshpande, Vahan Martisorian, Peter LaViolette, Adrienne Boire, Frances Chow, Kyle Hurth, Peter Fecci, Mark Shiroishi, and Rachel Eisenbarth
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
Control of breast to brain metastasis remains an urgent unmet clinical need. While chemotherapies are essential in reducing systemic tumor burden, they have also shown to promote non-brain metastatic invasiveness and drug-driven neurocognitive deficits through formation of neurofibrillary tangles (NFT) independently. Now, in this study we investigated the effect of chemotherapy on brain metastatic progression and promoting tumor-mediated NFT. Results show chemotherapies promote increased brain-barrier permeability and facilitate enhanced tumor infiltration, particularly through the blood-cerebrospinal fluid-barrier (BCSFB). This is attributed to increased expression of matrix metalloproteinase 9 (MMP9) which, in turn, mediates loss of Claudin-6 within the choroid plexus cells of the BCSFB. Importantly, increased MMP9 activity in the choroid epithelium following chemotherapy results in cleavage of Tau released from breast cancer cells. This cleaved Tau forms tumor-derived NFT that further destabilize the BCSFB. Our results underline for the first time the importance of the BCSFB as a vulnerable point of entry for brain-seeking tumor cells post-chemotherapy and indicate that tumor cells themselves contribute to Alzheimer’s-like tauopathy.
- Published
- 2022
- Full Text
- View/download PDF
9. PATH-22. CLINICAL FEATURES AND MOLECULAR CHARACTERIZATION OF LEPTOMENINGEAL DISEASE IN PATIENTS WITH HIGH GRADE GLIOMA
- Author
-
Madison Shoaf, Frances Chow, Joanne Xiu, Michael Glantz, Sonikpreet Aulakh, David Ashley, Eric S Lipp, Giselle Lopez, Ashley Sumrall, Phillip Walker, David Spetzler, Theodore Nicolaides, and Katherine B Peters
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
INTRODUCTION Leptomeningeal disease (LMD) is a challenging complication of high grade glioma (HGG) and critical questions remain unanswered regarding clinicopathologic risk factors, molecular associations, and optimal treatment. METHODS Patients with molecularly-profiled HGG (Caris Life Sciences; Phoenix, AZ) with LMD at two institutions were included. Medical records were reviewed for clinicopathological characteristics, treatment, and outcome. Kaplan-Meier estimates of patient survival were performed on censored data using Cox’s proportional hazard model. RESULTS 43 patients (male: 33, female: 10; median age: 56 years) were identified, comprising 41 grade 4 (glioblastoma: 38; gliosarcoma: 2; H3K27M diffuse midline glioma: 1) and 2 grade 3 tumors (astrocytoma: 1; pleomorphic xanthoastrocytoma: 1). LMD diagnosed at HGG diagnosis (n=18) versus recurrence (n=22) was associated with longer post-LMD survival [pLMD-OS: 15.3m vs. 4.8m, HR: 0.07, 95% CI: 0.02-0.29, p=0.0004] but similar overall survival [mOS: 15.3m vs. 12.3m; HR: 0.82; 95% CI: 0.36-1.85; p=0.63]. Pathology-diagnosed LMD (n=15) versus MRI-diagnosed LMD (n=26) was associated with longer post-LMD survival [pLMD-OS: 15.4m vs. 5.2m, HR: 14.9, 95% CI: 0.01-0.30, p=0.0004] but similar overall survival [mOS: 17.1m vs. 12.3m; HR: 0.66; 95% CI: 0.3-1.58; p=0.38]. Post-LMD survival was significantly prolonged for supratentorial (n=28) versus infratentorial/spinal (n=4) locations regardless of the diagnostic modality [pLMD-OS: 2.6m vs. 11.3m, HR: 14.4, 95% CI: 2.73-75.7, p=0.0017], and did not significantly differ between symptomatic (n=20) and asymptomatic (n=23) patients [pLMD-OS: 4.8m vs. 11.2m, HR: 1.75, 95% CI: 0.82-3.77, p=0.15). pTERT mutation (81%), EGFR amplification (43%), and MGMT methylation (33%) were prevalent but IDH1 mutation was rare (2.8%). Comparison with a separate glioblastoma cohort (n=1400) suggested more frequent amplification of CHIC2, MDM4, and KDR, higher mutation rates of RUNX1, APC, and RAD51C, colder tumor microenvironment (TME), and lower expression of immune checkpoint-related genes. CONCLUSIONS Clinicopathological characteristics affect post-LMD survival, and cohort comparison suggests molecular and TME differences in LMD-HGG tumors.
- Published
- 2022
- Full Text
- View/download PDF
10. CTNI-17. A MULTI-INSTITUTIONAL RANDOMIZED CLINICAL TRIAL COMPARING ASSAY - GUIDED CHEMOTHERAPY WITH PHYSICIAN-CHOICE TREATMENT FOR RECURRENT HIGH-GRADE GLIOMA (NCT03632135)
- Author
-
Tulika Ranjan, Soma Sengupta, Michael Glantz, Richard Green, Alexander Yu, Dawit Aregawi, Rekha Chaudary, Ricky Chen, Mario Zuccarello, Christine Lu-Emerson, Hugh Moulding, Neil Belman, Jon Glass, Aaron Mammoser, Mark Anderson, Nicholas Marko, Jason Schroeder, Steven Jubelirer, Frances Chow, Anthony Alberico, Candace Howard, Seth Lirette, Krista Denning, Jagan Valluri, and Pier Paolo Claudio
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
The presence of therapy-resistant cancer stem cells (CSCs) in recurrent high-grade glioma (HGG) patients contributes to poor clinical outcomes. The ChemoID functional anti-cancer assay targets cancer stem cells along with the bulk of the tumor cells. This trial aims to determine if ChemoID assay-guided treatment improves survival rates for recurrent HGG patients compared to the empirically physician-selected treatment. Patients with grade-III/IV recurrent glioma who failed standard of care (SOC) therapy were randomized (1:1) between two intervention groups. They received one of fourteen mono or combination chemotherapies based on the ChemoID assay or physician choice. The study met the primary outcome in the first interim analysis of 50 patients as per protocol. The ChemoID group had an improved survival rate (vs physician-choice). Median OS (mOS) was 12.5 months in the ChemoID group (95% CI, 10.2-14.7) vs 9 months in the physician-choice (95% CI, 4.2-13.8; log-rank P = .010). Mortality risk was lower in the ChemoID group (HR = 0.44; 95% CI, 0.24-0.81; P = .008). Median progression-free survival was 10.1 months in the ChemoID group vs 3.5 months in the physician choice (95% CI, 4.8-15.4 vs 1.9-5.1; log-rank < 0.001). Risk of progression was lower in the ChemoID group (HR = 0.25; 95% CI, 0.14-0.44; P < 0.001). The intention to treat (ITT) analysis of 78 patients showed substantially improved OS. The ChemoID group had a statistically significant longer median survival of 4.5 months. mOS was 12.0 months in the ChemoID group (95% CI, 10.8-13.2) vs 7.5 in the physician-choice group (95% CI, 3.5-11.5; log-rank P = .009). The ChemoID group had a decreased mortality risk (HR = 0.52; 95% CI, 0.24-0.81; P = .008). Compared with the physician-choice, the ChemoID group had a significantly longer OS in the ITT population. Our findings support that screening standard cytotoxic chemotherapies with a patient-specific anti-cancer assay improves survival outcomes in recurrent HGG patients.
- Published
- 2022
- Full Text
- View/download PDF
11. Abstract 3124: Identification of sub-clonal loss of heterozygosity (LOH) in glioblastomas analyzed with spatial transcriptomics
- Author
-
Michelle G. Webb, Frances Chow, Carmel McCullough, Bohan Zhang, Kyle Hurth, John Carpten, Gabriel Zada, and David W. Craig
- Subjects
Cancer Research ,Oncology - Abstract
We propose a method to leverage spatial transcriptomics and bioinformatic analysis to identify sub-clonal loss of heterozygosity that could otherwise be missed by standard bulk sequencing in glioblastoma tumors. Glioblastomas are highly invasive and aggressive brain tumors with a low five-year survival rate of less than 10%. Current treatment strategies are not sufficient for long term disease outcome, and innovative approaches to analyze glioblastoma are imperative for future improved options. Many factors contribute to difficulty of treatment, though one critical consideration is the diffuse nature and known intratumoral heterogeneity of this cancer. It is important to determine LOH heterogeneity, as this genomic alteration is associated with copy number variation and specific classifications of gliomas. Loss of heterozygosity is an irreversible deletion of DNA that is a common alteration in cancer. Application of spatial transcriptomics can increase our understanding of genomic complexity across a tissue, and unsupervised graph clustering can computationally separate spatial regions with similar gene expression patterns. Our method investigates spatial clusters individually at known heterozygous SNP sites to identify evidence for LOH with Bayesian inference and a hidden Markov model. Our analysis centers on the balance of reference (A) and non-reference (B) spatial transcriptomics read counts at SNPs of interest. B allele frequency is the number of reads aligned to the alternative allele divided by the total number of reads counted at the SNP. At a normal two-copy state at a heterozygous site, B allele frequency is ideally 50 percent. LOH can be identified as this value deviates toward 0 or 100, and copy number alterations impact this frequency. Our method utilizes Bayes factor values, or likelihood ratios between two models, using allele counts to provide support for either LOH or a heterozygous event. For our analysis, spatial transcriptomics BAMs are processed with a graph clustering algorithm and split into cluster-specific BAMs. We calculate allele count coverage for each cluster at pre-selected heterozygous SNP positions identified through germline exome sequencing. Bayes factor values are then calculated at each SNP, resulting in evidence for LOH or a heterozygous event. Chromosomal regions are segmented by a hidden Markov approach. Cumulative metrics for each segment are evaluated to determine a “state” label of heterozygous, LOH, or undefined. We tested our method on three WHO grade 4 IDH wild-type EGFRvIII positive fresh frozen glioblastoma tissue samples. Sub-clonal heterogeneity was identified in two samples and confirmed by bulk exome sequencing, bulk RNA sequencing, and analysis of initial clinical reports. Citation Format: Michelle G. Webb, Frances Chow, Carmel McCullough, Bohan Zhang, Kyle Hurth, John Carpten, Gabriel Zada, David W. Craig. Identification of sub-clonal loss of heterozygosity (LOH) in glioblastomas analyzed with spatial transcriptomics [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 3124.
- Published
- 2023
- Full Text
- View/download PDF
12. SYST-01 MULTI-INSTITUTIONAL RANDOMIZED TRIAL COMPARING CANCER STEM CELL-TARGETED VS PHYSICIAN-CHOICE TREATMENTS IN PATIENTS WITH RECURRENT HIGH-GRADE GLIOMAS (NCT03632135)
- Author
-
Soma Sengupta, Tulika Ranjan, Alexander Yu, Candace Howard, Ricky Chen, Rekha Chaudahary, Nicholas Marko, Darit Aregawi, Michael Glantz, Jon Glass, Richard Green, Christine Lu-Hemerson, Aaaron Mammoser, Hugh Moulding, Steven Jubelirer, Jason Schoroeder, Mark Anderson, Frances Chow, Seth Lirette, Krista Denning, Anthony Alberico, Jagan Valluri, and Pier Paolo Claudio
- Subjects
General Medicine - Abstract
BACKGROUND Clinical outcomes in patients with recurrent high-grade glioma (HGG) remain poor. Cancer stem cells (CSCs) have been implicated in metastasis, treatment resistance and recurrence of HHGs. We have shown in several clinical studies that anti-CSC-directed therapy provides benefits in many cancer types; however, this is the first report of a randomized clinical trial evaluating it for recurrent HGGs. OBJECTIVE Determine whether CSC-targeted cytotoxic agents selected by ChemoID assay-guided therapy improves survival in patients with recurrent HGG. DESIGN, SETTING, AND PARTICIPANTS In this parallel-group, randomized, phase-3 clinical trial, patients at 13 clinical sites in the USA with grade-III/IV recurrent glioma (2016 WHO guidelines) were randomized 1:1 to either ChemoID assay-guided therapy or physician-choice therapy, and then treated and followed until unacceptable toxic effects, hospice, or death. MAIN OUTCOMES AND MEASURES The primary endpoint was overall survival (OS). RESULTS Combined median follow-up was 9 months. Median OS (mOS) was 12.5 months (95% CI, 10.2-14.7) in the ChemoID assay-guided group vs 9 months (95% CI, 4.2-13.8) in the physician-choice group (log-rank P = .010). Risk of death was significantly lower in the ChemoID assay group (HR = 0.44; 95% CI, 0.24-0.81; P = .008). Median progression free survival (PFS) was 10.1 vs 3.5 months (95% CI, 4.8-15.4 vs 1.9-5.1) (HR, 0.25; 95% CI, 0.14-0.44; P < .001). CONCLUSIONS AND RELEVANCE Primary endpoint was met in this randomized clinical trial. The mOS was 3.5 months longer in the ChemoID assay-guided group vs the physician-choice group.
- Published
- 2022
- Full Text
- View/download PDF
13. 905: POST-INTUBATION PARTIAL PRESSURE OF O2/FRACTION OF INSPIRED O2 AND IN-HOSPITAL MORTALITY IN COVID-19
- Author
-
Claire Baldauf, Frances Chow, Jonathan Warus, Yuding Wang, Elizabeth Kermgard, Elizabeth Burner, Ghazal Kabbach, Dafang Chen, and Keith Killu
- Subjects
Critical Care and Intensive Care Medicine - Published
- 2022
- Full Text
- View/download PDF
14. BSCI-12 BREAST TO BRAIN METASTASIS IS EXACERBATED WITH CHEMOTHERAPY THROUGH BLOOD-CEREBRAL SPINAL FLUID-BARRIER AND INDUCES ALZHEIMER’S-LIKE PATHOLOGY
- Author
-
Josh Neman, Behnaz Saatian, Robert Herrera, Vahan Martirosian, Rachel Eisenbarth, Mukund Iyer, Alex Julian, Allison Lowman, Pete LaViolette, Jan Remsik, Adrienne Boire, E Sankey, Peter E Fecci, Mark Shiroishi, Frances Chow, and Kyle Hurth
- Subjects
General Medicine - Abstract
Control of breast to brain metastasis remains an urgent unmet clinical need. While chemotherapies are essential in reducing systemic tumor burden, they have also shown to promote non-brain metastatic invasiveness and drug-driven neurocognitive deficits through formation of neurofibrillary tangles (NFT) independently. Now, in this study we investigated the effect of chemotherapy on brain metastatic progression and promoting tumor-mediated NFT. Results show chemotherapies promote increased brain-barrier permeability and facilitate enhanced tumor infiltration, particularly through the blood-cerebrospinal fluid-barrier (BCSFB). This is attributed to increased expression of matrix metalloproteinase 9 (MMP9) which, in turn, mediates loss of Claudin-6 within the choroid plexus cells of the BCSFB. Importantly, increased MMP9 activity in the choroid epithelium following chemotherapy results in cleavage of Tau released from breast cancer cells. This cleaved Tau forms tumor-derived NFT that further destabilize the BCSFB. Our results underline for the first time the importance of the BCSFB as a vulnerable point of entry for brain-seeking tumor cells post-chemotherapy and indicate that tumor cells themselves contribute to Alzheimer’s-like tauopathy.
- Published
- 2022
- Full Text
- View/download PDF
15. Utilization of Discarded Surgical Tissue from Ultrasonic Aspirators to Establish Patient-Derived Metastatic Brain Tumor Cells: A Guide from the Operating Room to the Research Laboratory
- Author
-
Josh Neman, Casey A. Jarvis, Steven L. Giannotta, Vahan Martirosian, Krutika Deshpande, Edith Yuan, Frank J. Attenello, Frances Chow, Thomas C. Chen, Michelle Lin, and Gabriel Zada
- Subjects
Pathology ,medicine.medical_specialty ,Operating Rooms ,Ultrasonic Therapy ,Population ,Tumor resection ,Brain tumor ,Health Informatics ,General Biochemistry, Genetics and Molecular Biology ,Article ,medicine ,Humans ,Ultrasonics ,General Pharmacology, Toxicology and Pharmaceutics ,education ,Protocol (science) ,education.field_of_study ,General Immunology and Microbiology ,business.industry ,Brain Neoplasms ,General Neuroscience ,medicine.disease ,Medical Laboratory Technology ,Metastatic brain tumor ,Cell culture ,Cancer cell ,business ,Laboratories ,Brain metastasis - Abstract
Patient-derived cells from surgical resections are of paramount importance to brain tumor research. It is well known that there is cellular and microenvironmental heterogeneity within a single tumor mass. Thus, current established protocols for propagating tumor cells in vitro are limiting because resections obtained from conventional singular samples limit the diversity in cell populations and do not accurately model the heterogeneous tumor. Utilization of discarded tissue obtained from cavitron ultrasonic surgical aspirator (CUSA) of the whole tumor mass allows for establishing novel cell lines in vitro from the entirety of the tumor, thereby creating an accurate representation of the heterogeneous population of cells originally present in the tumor. Furthermore, while others have described protocols for establishing patient tumor lines once tissue has arrived in the research lab, a primer from the operating room (OR) to the research lab has not been described before. This is integral, as basic research scientists need to understand the surgical environment of the OR, including the methods utilized to obtain a patient's tumor resection, in order to more accurately model cancer biology in laboratory. © 2021 Wiley Periodicals LLC. Basic Protocol 1: Establishment of brain tumor cell lines from patient-derived CUSA samples: processing brain tumor sample from the OR to the lab Support Protocol 1: Sterilization of microsurgical tools in preparation for dissection Support Protocol 2: Collagen coating of tissue culture flasks Basic Protocol 2: Selection of tumor cells in vitro Support Protocol 3: FACS sorting tumor sample to isolate cancer cells from heterogeneous cell population.
- Published
- 2021
16. Resolution of tissue signatures of therapy response in patients with recurrent GBM treated with neoadjuvant anti-PD1
- Author
-
Timothy F. Cloughesy, Yue Lu, James R. Heath, Frances Chow, Richard Everson, Robert M. Prins, Michael T. Tetzlaff, Jennifer A. Wargo, Alphonsus H. C. Ng, Rohit Thakur, and Beth A. Helmink
- Subjects
0301 basic medicine ,Male ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,General Physics and Astronomy ,CD8-Positive T-Lymphocytes ,urologic and male genital diseases ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Tumor Microenvironment ,Medicine ,Cytotoxic T cell ,CTLA-4 Antigen ,Melanoma ,Immune Checkpoint Inhibitors ,Neoadjuvant therapy ,Cancer ,Multidisciplinary ,Tumor ,Brain Neoplasms ,Middle Aged ,female genital diseases and pregnancy complications ,Neoadjuvant Therapy ,Treatment Outcome ,Immunological ,Nivolumab ,Local ,030220 oncology & carcinogenesis ,Female ,Systems biology ,Cancer microenvironment ,Adult ,Science ,Immunology ,Antineoplastic Agents ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Immune system ,Rare Diseases ,Biomarkers, Tumor ,Humans ,neoplasms ,Aged ,Tumor microenvironment ,urogenital system ,business.industry ,Neurosciences ,General Chemistry ,medicine.disease ,Ipilimumab ,Immune checkpoint ,nervous system diseases ,Blockade ,Brain Disorders ,Brain Cancer ,030104 developmental biology ,Neoplasm Recurrence ,Cancer research ,Neoplasm Recurrence, Local ,business ,Glioblastoma ,CD8 ,Biomarkers - Abstract
The response of patients with recurrent glioblastoma multiforme to neoadjuvant immune checkpoint blockade has been challenging to interpret due to the inter-patient and intra-tumor heterogeneity. We report on a comparative analysis of tumor tissues collected from patients with recurrent glioblastoma and high-risk melanoma, both treated with neoadjuvant checkpoint blockade. We develop a framework that uses multiplex spatial protein profiling, machine learning-based image analysis, and data-driven computational models to investigate the pathophysiological and molecular factors within the tumor microenvironment that influence treatment response. Using melanoma to guide the interpretation of glioblastoma analyses, we interrogate the protein expression in microscopic compartments of tumors, and determine the correlates of cytotoxic CD8+ T cells, tumor growth, treatment response, and immune cell-cell interaction. This work reveals similarities shared between glioblastoma and melanoma, immunosuppressive factors that are unique to the glioblastoma microenvironment, and potential co-targets for enhancing the efficacy of neoadjuvant immune checkpoint blockade., The response to neoadjuvant immune checkpoint blockade (ICB) in patients with recurrent gliolastoma multiforme (GBM) has been challenging to interpret. Here the authors develop a tumor analysis framework that reveals molecular similarities between GBM and melanoma and unique patterns of immunosuppression in GBM indicating potential co-targets for neoadjuvant ICB.
- Published
- 2021
17. CTNI-17. A PHASE 1 WITH DOSE EXPANSION/PHASE 2 STUDY OF SELINEXOR IN COMBINATION WITH STANDARD OF CARE THERAPY FOR NEWLY DIAGNOSED OR RECURRENT GLIOBLASTOMA
- Author
-
Howard Colman, Frances Chow, Jatin P. Shah, Gregory Mundy, Andrew B. Lassman, Rebecca Harrison, Yasaman Damestani, Yang Liu, Vyshak Venur, Warren P. Mason, Nicholas Butowski, Minesh P. Mehta, Paul Duic, Sharon Tamir, Michael Schulder, John A. Boockvar, Kai Li, Yazmin Odia, Patrick Y. Wen, Erin M. Dunbar, Samuel Goldlust, Sharon Shacham, Scott R. Plotkin, Eric Sbar, and Priya Kumthekar
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Standard of care ,Temozolomide ,Bevacizumab ,business.industry ,Recurrent glioblastoma ,O-6-methylguanine-DNA methyltransferase ,Lomustine ,26th Annual Meeting & Education Day of the Society for Neuro-Oncology ,medicine.disease ,Glioma ,Internal medicine ,Troponin I ,medicine ,Neurology (clinical) ,business ,medicine.drug - Abstract
BACKGROUND Glioblastoma (GBM) is the most common and aggressive primary brain tumor with median overall survival of 15 months and 5-7 months for patients with newly diagnosed or recurrent disease (nGBM or rGBM), respectively. Selinexor is a first-in-class, oral, selective inhibitor of nuclear export which blocks exportin 1 (XPO1), forcing the nuclear retention and reactivation of tumor suppressor proteins, ultimately causing cancer cell death. Increased XPO1 expression in gliomas is associated with higher pathological grade and poorer prognosis. Consistent with these data, selinexor inhibited tumor growth and prolonged survival in an animal model of GBM. Importantly, selinexor showed encouraging intra-tumoral penetration and single-agent efficacy in rGBM (KING study). The current trial tests the hypothesis that adding selinexor to standard therapy will improve clinical outcomes for patients with nGBM or rGBM. METHODS To facilitate the successful development of new therapies, consensus recommendations are to use biomarker enrichment and flexible design that allows expansion of promising cohorts. Accordingly, this phase 1a dose finding study is followed by a phase 1b dose expansion (and ultimately by a 1:1 randomized phase 2 efficacy exploration trial) to independently evaluate: radiation + selinexor in nGBM with unmethylated MGMT promoter (Arm A), radiation + temozolomide + selinexor for nGBM with methylated MGMT promoter (Arm B), and lomustine + selinexor in rGBM (Arm C). Bevacizumab or TTField + selinexor in rGBM (Arms D & E, respectively) are being considered. The Phase 1a primary endpoint is maximum tolerated dose/recommended phase 2 dose. The phase 1b primary endpoint is PFS at 3 months against historic controls. The phase 1b dose expansion is included to evaluate preliminary efficacy before launching into a randomized phase 2 trial. Patient quality of life during the trial will be objectively measured using digital devices (e.g. smartwatch/customized smartphone app). We are currently enrolling patients nationwide. Clinical Trial Registration number: NCT04421378.
- Published
- 2021
- Full Text
- View/download PDF
18. Complementary and Alternative Medicine for the Treatment of Gliomas: Scoping Review of Clinical Studies, Patient Outcomes, and Toxicity Profiles
- Author
-
Frances Chow, Tyler Cardinal, Eliza M. Kellman, Ben A. Strickland, Gabriel Zada, Martin J. Rutkowski, Frank J. Attenello, Andrew Brunswick, Dhiraj J. Pangal, and Hans Baertsch
- Subjects
Oncology ,Complementary Therapies ,medicine.medical_specialty ,medicine.medical_treatment ,Recurrent Glioma ,03 medical and health sciences ,0302 clinical medicine ,Mistletoe extract ,Carbogen ,Internal medicine ,Glioma ,medicine ,Humans ,business.industry ,Brain Neoplasms ,Evidence-based medicine ,medicine.disease ,Treatment Outcome ,030220 oncology & carcinogenesis ,Toxicity ,Surgery ,Carbogen Breathing ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,Ketogenic diet - Abstract
Introduction Complementary and alternative medicine (CAM) are highly used among those diagnosed with glioma. Further research is warranted, however, as it remains important to clearly delineate CAM practices that are unproven, disproven, or promising for future research and implementation. Methods A systematic review was conducted to identify all articles that investigated the effect of any CAM therapy on survival of patients with newly diagnosed or recurrent glioma. Results Eighteen papers and 4 abstracts pertaining to the effects of ketogenic diet (4), antioxidants (3), hyperbaric oxygen (4), cannabinoids (2), carbogen and nicotinamide (3), mistletoe extract (2), hypocupremia and penicillamine (1), and overall CAM use (3) on overall and progression-free survival in patients with low- and high-grade glioma were identified (Levels of Evidence I-IV). Ketogenic diets, hyperbaric oxygen therapy, and cannabinoids appear to be safe and well tolerated by patients; preliminary studies demonstrate tumor response and increased progression-free survival and overall survival when combined with standard of care therapies. Antioxidant usage exhibit mixed results perhaps associated with glioma grade with greater effect on low-grade gliomas; vitamin D intake was associated with prolonged survival. Conversely, carbogen breathing and hypocupremia were found to have no effect on the survival of patients with glioma, with associated significant toxicity. Most modalities under the CAM umbrella have not been appropriately studied and require further investigation. Conclusions Despite widespread use, Level I or II evidence for CAM for the treatment of glioma is lacking, representing future research directions to optimally counsel and treat glioma patients.
- Published
- 2021
19. Unique challenges for glioblastoma immunotherapy-discussions across neuro-oncology and non-neuro-oncology experts in cancer immunology. Meeting Report from the 2019 SNO Immuno-Oncology Think Tank
- Author
-
Frances Chow, Michael Platten, Wolfgang Wick, Benjamin M. Ellingson, Pavlina Chuntova, David A. Reardon, Christine E. Brown, Derek A. Wainwright, Timothy F. Cloughesy, Ming Li, Michael Lim, Duane Mitchell, Yvonne Y. Chen, Hideho Okada, Ronald A. DePinho, Jay A. Berzofsky, James R. Heath, Kim Margolin, Payal Watchmaker, Masaki Terabe, Robert M. Prins, Peter E. Fecci, William Timmer, Evan W. Newell, Aaron Diaz, Mildred Galvez, E. Antonio Chiocca, Joseph F. Costello, E. John Wherry, Noriyuki Kasahara, Patrick Y. Wen, Linda M. Liau, Amy B. Heimberger, Christel Herold-Mende, and Gavin P. Dunn
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Neuro oncology ,Oncology and Carcinogenesis ,Meeting Report ,Medical Oncology ,Vaccine Related ,Rare Diseases ,Cancer immunotherapy ,Internal medicine ,Neoplasms ,medicine ,Tumor Microenvironment ,Humans ,Oncology & Carcinogenesis ,Cancer immunology ,Cancer ,Tumor microenvironment ,immunosuppression ,business.industry ,Brain Neoplasms ,glioblastoma ,Neurosciences ,clinical trial ,Immunotherapy ,medicine.disease ,Immune checkpoint ,Brain Disorders ,Clinical trial ,Brain Cancer ,conference report ,Orphan Drug ,Good Health and Well Being ,Immunization ,Neurology (clinical) ,immunotherapy ,business ,Glioblastoma - Abstract
Cancer immunotherapy has made remarkable advances with over 50 separate Food and Drug Administration (FDA) approvals as first- or second-line indications since 2015. These include immune checkpoint blocking antibodies, chimeric antigen receptor-transduced T cells, and bispecific T-cell–engaging antibodies. While multiple cancer types now benefit from these immunotherapies, notable exceptions thus far include brain tumors, such as glioblastoma. As such, it seems critical to gain a better understanding of unique mechanistic challenges underlying the resistance of malignant gliomas to immunotherapy, as well as to acquire insights into the development of future strategies. An Immuno-Oncology Think Tank Meeting was held during the 2019 Annual Society for Neuro-Oncology Scientific Conference. Discussants in the fields of neuro-oncology, neurosurgery, neuro-imaging, medical oncology, and cancer immunology participated in the meeting. Sessions focused on topics such as the tumor microenvironment, myeloid cells, T-cell dysfunction, cellular engineering, and translational aspects that are critical and unique challenges inherent with primary brain tumors. In this review, we summarize the discussions and the key messages from the meeting, which may potentially serve as a basis for advancing the field of immune neuro-oncology in a collaborative manner.
- Published
- 2021
20. Phase I trial of intranasal NEO100, highly purified perillyl alcohol, in adult patients with recurrent glioblastoma
- Author
-
Rose Lai, Clovis Orlando da Fonseca, Frances Chow, Kyle Hurth, Lynne Taylor, Axel H. Schönthal, David M. Peereboom, Vincent F Simmon, Anna Mathew, Naveed Wagle, Thomas C. Chen, and Steven P. Howard
- Subjects
medicine.medical_specialty ,Dose ,Clinical Investigations ,Phases of clinical research ,O6-methylguanine-DNA methyltransferase ,Gastroenterology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,isocitrate dehydrogenase 1 ,Internal medicine ,Glioma ,medicine ,AcademicSubjects/MED00300 ,Dosing ,Adverse effect ,business.industry ,Perillyl alcohol ,intranasal ,perillyl alcohol ,medicine.disease ,Clinical trial ,chemistry ,030220 oncology & carcinogenesis ,Nasal administration ,AcademicSubjects/MED00310 ,business ,030217 neurology & neurosurgery ,recurrent glioblastoma - Abstract
Background Better treatments for glioblastoma (GBM) patients, in particular in the recurrent setting, are urgently needed. Clinical trials performed in Brazil indicated that intranasal delivery of perillyl alcohol (POH) might be effective in this patient group. NEO100, a highly purified version of POH, was current good manufacturing practice (cGMP) manufactured to evaluate the safety and efficacy of this novel approach in a Phase I/IIa clinical trial in the United States. Methods A total of 12 patients with recurrent GBM were enrolled into Phase I of this trial. NEO100 was administered by intranasal delivery using a nebulizer and nasal mask. Dosing was 4 times a day, every day. Four cohorts of 3 patients received the following dosages: 96 mg/dose (384 mg/day), 144 mg/dose (576 mg/day), 192 mg/dose (768 mg/day), and 288 mg/dose (1152 mg/day). Completion of 28 days of treatment was recorded as 1 cycle. Adverse events were documented, and radiographic response via Response Assessment in Neuro-Oncology (RANO) criteria was evaluated every 2 months. Progression-free and overall survival were determined after 6 and 12 months, respectively (progression-free survival-6 [PFS-6], overall survival-12 [OS-12]). Results Intranasal NEO100 was well tolerated at all dose levels and no severe adverse events were reported. PFS-6 was 33%, OS-12 was 55%, and median OS was 15 months. Four patients (33%), all of them with isocitrate dehydrogenase 1 (IDH1)-mutant tumors, survived >24 months. Conclusion Intranasal glioma therapy with NEO100 was well tolerated. It correlated with improved survival when compared to historical controls, pointing to the possibility that this novel intranasal approach could become useful for the treatment of recurrent GBM.
- Published
- 2021
21. Abstract CT224: Multi-institutional randomized phase-3 trial comparing cancer stem cell-targeted vs physician-choice treatments in patients with recurrent high-grade gliomas (NCT03632135)
- Author
-
Tulika Ranjan, Soma Sengupta, Alexander Yu, Candace M. Howard, Ricky Chen, Rekha Chaudhary, Nicholas Marko, Dawit Aregawi, Michael Glantz, Jon Glass, Richard M. Green, Christine Lu-Emerson, Aaron Mammoser, Hugh Moulding, Steven Jubelirer, Jason Schroeder, Mark Anderson, Frances Chow, Seth Lirette, Krista Denning, Anthony Alberico, Jagan Valluri, and Pier Paolo Claudio
- Subjects
Cancer Research ,Oncology - Abstract
Background: Clinical outcomes in patients with recurrent high-grade glioma (HGG) remain poor. Cancer stem cells (CSCs) have been implicated in metastasis, treatment resistance and recurrence of HHGs. We have shown in several clinical studies that anti-CSC-directed therapy provides benefits in many cancer types; however, this is the first report of a randomized clinical trial evaluating it for recurrent HGGs. Objective: Determine whether CSC-targeted cytotoxic agents selected by ChemoID assay-guided therapy improves survival in patients with recurrent HGG. Design, Settings, and Participants: In this parallel-group, randomized, phase-3 clinical trial, patients at 13 clinical sites in the USA with grade-III/IV recurrent glioma (2016 WHO guidelines) were randomized 1:1 to either ChemoID assay-guided therapy or physician-choice therapy, and then treated and followed until unacceptable toxic effects, hospice, or death. Main Outcomes and Measures: The primary endpoint was overall survival (OS). Results: Combined median follow-up was 9 months. Median OS (mOS) was 12.5 months (95% CI, 10.2-14.7) in the ChemoID assay-guided group vs 9 months (95% CI, 4.2-13.8) in the physician-choice group (log-rank P = .010). Risk of death was significantly lower in the ChemoID assay group (HR = 0.44; 95% CI, 0.24-0.81; P = .008). Median progression free survival (PFS) was 10.1 vs 3.5 months (95% CI, 4.8-15.4 vs 1.9-5.1) (HR, 0.25; 95% CI, 0.14-0.44; P < .001). Conclusions and Relevance: Primary endpoint was met in this randomized clinical trial. The mOS was 3.5 months longer in the ChemoID assay-guided group vs the physician-choice group demonstrating the clinical advantage of treating HGG patients using CSC personalized therapy. Citation Format: Tulika Ranjan, Soma Sengupta, Alexander Yu, Candace M. Howard, Ricky Chen, Rekha Chaudhary, Nicholas Marko, Dawit Aregawi, Michael Glantz, Jon Glass, Richard M. Green, Christine Lu-Emerson, Aaron Mammoser, Hugh Moulding, Steven Jubelirer, Jason Schroeder, Mark Anderson, Frances Chow, Seth Lirette, Krista Denning, Anthony Alberico, Jagan Valluri, Pier Paolo Claudio. Multi-institutional randomized phase-3 trial comparing cancer stem cell-targeted vs physician-choice treatments in patients with recurrent high-grade gliomas (NCT03632135) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr CT224.
- Published
- 2022
- Full Text
- View/download PDF
22. IMMU-20. SINGLE-CELL RNASEQ OF TUMOR INFILTRATING IMMUNE CELLS FROM NEOADJUVANT ANTI-PD1 TREATED GBM PATIENTS REVEALS GLOBAL TRANSCRIPTIONAL CHANGES AND IMMUNOSUPPRESSIVE ADAPTIVE RESPONSES BY MYELOID CELLS
- Author
-
Lu Sun, Joey Orpilla, Steven J. Bensinger, Frances Chow, Jeremy Reynoso, Robert M. Prins, David Nathanson, Linda M. Liau, Carolina Chavez, Jenny Kienzler, Timothy F. Cloughesy, Mildred Galvez, Richard Everson, Aaron Mochizuki, Willy Hugo, William H. Yong, and Alexander Lee
- Subjects
Cancer Research ,medicine.anatomical_structure ,Immune system ,Oncology ,Cell ,Myeloid cells ,Immunology ,Cancer research ,medicine ,Neurology (clinical) ,Biology ,Anti pd1 - Abstract
INTRODUCTION Neoadjuvant anti-PD1 therapy (neo-aPD1) was previously shown to significantly increase the survival of recurrent glioblastoma patients in a small randomized clinical trial. However, neo-aPD1 alone was not curative so defining the limitations of neo-aPD1 and discovering where other immunotherapies can be used alongside neo-aPD1 is needed. METHODS To understand how immune cells in the tumor microenvironment change with neo-aPD1, we used single-cell RNAsequencing to analyze cells from 27 glioma patients (n = 105,143 cells) of which 9 patients had received neo-aPD1 (n = 33,325 cells). Using unsupervised clustering and pseudotime trajectory analysis, we characterized the transcriptional changes within immune cells and how these populations changed with therapy. RESULTS We defined the immune landscape of the glioblastoma tumor microenvironment. Compared to no immunotherapy treatment, neo-aPD1 significantly increased the ratio of T cells to myelo-monocytic cells and led to significant increases in the effector and memory T cell populations but no significant changes in myeloid cell composition. Our differential gene expression analysis of the myeloid compartment showed significant increases in interferon-γ-responsive genes and down-regulation of genes associated with M2 macrophages and MDSCs, suggestive that neo-aPD1 influences the transcriptional profile of myeloid cells in the tumor microenvironment. Interestingly, our psuedotime trajectory analysis showed that neo-aPD1 was associated with cells expressing both lymphoid and myeloid-related genes, which we theorized to actually be lymphoid-myeloid cell doublets caused by increased interactions between myeloid and lymphoid cells. These doublets were highly enriched in MHC I and II, macrophage, T cell, and T cell activation and exhaustion genes indicating that neo-aPD1 may result in some adaptive immunosuppressive mechanism by increasing these interactions. This could explain why neo-aPD1 alone is not curative for glioblastoma patients. CONCLUSIONS In total, neoadjuvant anti-PD1 therapy enhances effector T cell activity, but may concomitantly induce adaptive resistance mediated by myeloid cells in glioblastoma.
- Published
- 2020
23. Comorbid depression in surgical cancer patients associated with non-routine discharge and readmission
- Author
-
Edith Yuan, Casey A. Jarvis, Phillip A. Bonney, Li Ding, Frank J. Attenello, Frances Chow, William J. Mack, Gabriel Zada, and Anthony W. Kim
- Subjects
Adult ,Male ,medicine.medical_specialty ,Databases, Factual ,Skilled Nursing ,Intermediate Care Facility ,Patient Readmission ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Sex Factors ,Risk Factors ,Home health ,Internal medicine ,Neoplasms ,Outcome Assessment, Health Care ,medicine ,Prevalence ,Humans ,030212 general & internal medicine ,Depression (differential diagnoses) ,Aged ,Retrospective Studies ,Depressive Disorder ,business.industry ,Cancer ,Middle Aged ,medicine.disease ,Primary cancer ,Primary tumor ,Patient Discharge ,Oncology ,030220 oncology & carcinogenesis ,Surgery ,Female ,business - Abstract
To characterize the rates of depression across primary cancer sites, and determine the effects of comorbid depression among surgical cancer patients on established quality of care indicators, non-routine discharge and readmission.Patients undergoing surgical resection for cancer were selected from the Nationwide Readmissions Database (2010-2014). Multivariable analysis adjusted for patient and hospital level characteristics to ascertain the effect of depression on post-operative outcomes and 30-day readmission rates. Non-routine discharge encompasses discharge to skilled nursing, inpatient rehabilitation, and intermediate care facilities, as well as discharge home with home health services.Among 851,606 surgically treated cancer patients, 8.1% had a comorbid diagnosis of depression at index admission (n = 69,174). Prevalence of depression was highest among patients with cancer of the brain (10.9%), female genital organs (10.9%), and lung (10.5%), and lowest among those with prostate cancer (4.9%). Depression prevalence among women (10.9%) was almost twice that of men (5.7%). Depression was associated with non-routine discharge after surgery (OR 1.20, CI:1.18-1.23, p 0.0001*) and hospital readmission within 30 days (OR 1.12, CI:1.09-1.15, p 0.001*).Rates of depression vary amongst surgically treated cancer patients by primary tumor site. Comorbid depression in these patients is associated with increased likelihood of non-routine discharge and readmission.
- Published
- 2020
24. Treatment at Safety-Net Hospitals Is Associated with Delays in Coil Embolization in Patients with Subarachnoid Hemorrhage
- Author
-
Daniel A. Donoho, Arun P. Amar, Ian A. Buchanan, Frances Chow, William J. Mack, Arati Patel, Frank J. Attenello, and Li Ding
- Subjects
Adult ,Male ,medicine.medical_specialty ,Multivariate analysis ,Subarachnoid hemorrhage ,Databases, Factual ,medicine.medical_treatment ,Psychological intervention ,Article ,Time-to-Treatment ,03 medical and health sciences ,0302 clinical medicine ,Aneurysm ,medicine ,Humans ,030212 general & internal medicine ,Healthcare Disparities ,Aged ,Coil embolization ,Medically Uninsured ,Endovascular coiling ,Medicaid ,business.industry ,Endovascular Procedures ,Middle Aged ,Subarachnoid Hemorrhage ,medicine.disease ,Embolization, Therapeutic ,United States ,Quartile ,Multivariate Analysis ,Emergency medicine ,Female ,Surgery ,Neurology (clinical) ,business ,Safety-net Providers ,030217 neurology & neurosurgery - Abstract
INTRODUCTION: Successful endovascular management of aneurysmal subarachnoid hemorrhage (aSAH) requires timely access to significant resources. Prior studies suggest an association between time to treatment and patient outcome. Patients treated at safety- net hospitals are thought to be particularly vulnerable to disparities in access to interventions that require substantial technological resources. We hypothesize that patients treated at safety-net hospitals are at greater risk for delayed access to endovascular treatment. MATERIALS AND METHODS: Adults undergoing endovascular coiling procedures between 2002–2011 in the Nationwide Inpatient Sample were included. Hospitals in the quartile with the highest proportion of Medicaid or uninsured patients were defined as safety-net hospitals. A multivariable model including patient and hospital-level factors was constructed to permit analysis of delays in endovascular treatment (defined as time to treatment greater than 3 days). RESULTS: Analysis included 7,109 discharges of patients with aSAH undergoing endovascular coil embolization procedures from 2002–2011. The median time to coil embolization in all patients was 1 day; 10.1% of patients waited more than three days until treatment. In multivariable analysis, patients treated at safety-net hospitals were more likely to have a prolonged time to coil embolization (OR 1.32, p
- Published
- 2018
- Full Text
- View/download PDF
25. TAMI-01. NEOADJUVANT PD-1 ANTIBODY BLOCKADE REMODELS THE IMMUNE MICROENVIRONMENT OF METASTATIC BRAIN TUMORS
- Author
-
Won Kim, Frances Chow, Richard Everson, William H. Yong, Lu Sun, Alexander Lee, Jenny Kienzler, Willy Hugo, Carolina Chavez, and Robert M. Prins
- Subjects
Cancer Research ,Oncology ,biology ,business.industry ,Immune microenvironment ,biology.protein ,Cancer research ,Medicine ,Tumor Microenvironment/Angiogenesis/Metabolism/Invasion ,Neurology (clinical) ,Antibody ,business ,Blockade - Abstract
Brain metastases (BM) commonly arise in patients with melanoma, lung, and breast cancer. Currently, there are limited options for GBM and BM patients who have failed the first-line standard treatment, underscoring the importance of developing new therapeutic strategies. Last year, we and other groups evaluated the neoadjuvant timing of anti-PD-1 checkpoint blockade therapy in recurrent GBM (rGBM) patients, which resulted in a modest survival benefit. In light of the known effectiveness of anti-PD-1 as a systemic therapy to control melanoma and non-small cell lung cancer BM, we set out to study the anti-tumor immune response of BM patients to anti-PD-1 in the neoadjuvant setting. We posited that neoadjuvant anti-PD-1 in patients with BM would result in a stronger antitumoral immune response, which could be quantified at the single cell level. To test this, we made use of contemporary single cell techniques, including multiplex immunofluorescence, time-of-flight mass cytometry (CyTOF) and single-cell RNA sequencing (scRNAseq), to characterize the intratumoral immune cell populations and their transcriptomic profiles. We found that neoadjuvant anti-PD-1 significantly increased the number of tumor infiltrating T lymphocytes in BM compared to rGBM (2.5 fold in BM, p= 0.02 vs. 1.4 fold in rGBM, p= 0.19). Multiplex immunofluorescence analysis of T cells in BM samples revealed a change from T cell exclusion to a diffusely infiltrating phenotype after anti-PD-1 treatment. Importantly, BM showed a higher fraction of effector/cytotoxic T cells compared to rGBM (7.3% vs. 0.9% of lymphoid cells, p= 0.005) and anti-PD-1 further enhanced this population. In the myeloid compartment of BM, neoadjuvant anti-PD-1 increased the frequency of HLA-DR+CD206- M1-like macrophages, implicating a pro-inflammatory microenvironment. In summary, our study delineated the immune cell subtypes altered by neoadjuvant anti-PD-1 and offers insights into new combination therapies that can help understand the clinical efficacy of immunotherapy for BM and GBM patients.
- Published
- 2020
- Full Text
- View/download PDF
26. Targeted next-generation sequencing of 565 neuro-oncology patients at UCLA: A single-institution experience
- Author
-
Stephanie Pan, Thomas J Lai, Matthew Ji, Frances Chow, Sean T Pianka, Phioanh L. Nghiemphu, Bryan Kevan, Linda M. Liau, Devin N Reeh, Christopher D Cox, Nhung T Nguyen, Regina Liu, Tie Li, William H. Yong, Timothy F. Cloughesy, Albert Lai, Blaine S C Eldred, Donna Molaie, and Gang Li
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Concordance ,PALB2 ,Clinical Investigations ,DNA sequencing ,03 medical and health sciences ,Rare Diseases ,0302 clinical medicine ,targeted next-generation sequencing ,Clinical Research ,Internal medicine ,Glioma ,glioma ,medicine ,CNS TUMORS ,Medical diagnosis ,Cancer ,screening and diagnosis ,Proportional hazards model ,business.industry ,Neurosciences ,glioblastoma ,CNS tumors ,medicine.disease ,Brain Disorders ,4.1 Discovery and preclinical testing of markers and technologies ,3. Good health ,Brain Cancer ,Detection ,Good Health and Well Being ,030104 developmental biology ,030220 oncology & carcinogenesis ,genomic profiling ,business ,4.2 Evaluation of markers and technologies ,Glioblastoma - Abstract
Background Targeted next-generation sequencing (NGS) is frequently obtained at the University of California, Los Angeles (UCLA) for clinical characterization of CNS tumors. In this study, we describe the diagnostic reliability of the Foundation Medicine (FM) targeted NGS platform and its ability to explore and identify tumor characteristics of prognostic significance in gliomas. Methods Neuro-oncology patients seen at UCLA who have received FM testing between August 2012 and March 2019 were included in this study, and all mutations from FM test reports were recorded. Initial tumor diagnoses and diagnostic markers found via standard clinical methods were obtained from pathology reports. With overall and progression-free survival data, elastic net regularized Cox regression and Cox proportional hazards models were used to determine whether any mutations of unknown significance detected by FM could predict patient outcome in glioblastoma (GBM). Results Six hundred and three samples tested by FM from 565 distinct patients were identified. Concordance of diagnostic markers was high between standard clinical testing methods and FM. Oligodendroglial markers detected via FM were highly correlated with 1p19q codeletion in IDH mutated gliomas. FM testing of multiple tumor samples from the same patient demonstrated temporal and spatial mutational heterogeneity. Mutations in BCORL1, ERBB4, and PALB2, which are mutations of unknown significance in GBM, were shown to be statistically significant in predicting patient outcome. Conclusions In our large cohort, we found that targeted NGS can both reliably and efficiently detect important diagnostic markers in CNS tumors.
- Published
- 2020
27. Amyloid Beta Oligomers Target to Extracellular and Intracellular Neuronal Synaptic Proteins in Alzheimer's Disease
- Author
-
Frances Chow, William L. Klein, Yang Zhang, Yu Ding, Shanshan Wang, Kirsten L. Viola, Xunle Zhang, Carol F. Lippa, Yuesong Gong, and Jiahui Zhao
- Subjects
0301 basic medicine ,Amyloid beta ,synGap ,soluble Aβ oligomers ,antibody to soluble Aβ oligomers ,DNA-binding protein ,lcsh:RC346-429 ,Synapse ,03 medical and health sciences ,0302 clinical medicine ,synapse ,Extracellular ,lcsh:Neurology. Diseases of the nervous system ,Original Research ,biology ,Chemistry ,Alzheimer's disease ,Ligand (biochemistry) ,Cell biology ,Blot ,030104 developmental biology ,postsynaptic density ,Shank3 ,Neurology ,biology.protein ,Neurology (clinical) ,α3-Na/K-ATPase ,Postsynaptic density ,030217 neurology & neurosurgery ,Intracellular - Abstract
Introduction: β-Amyloid protein (Aβ) putatively plays a seminal role in synaptic loss in Alzheimer's disease (AD). While there is no consensus regarding the synaptic-relevant species of Aβ, it is known that Aβ oligomers (AβOs) are noticeably increased in the early stages of AD, localizing at or within the synapse. In cell and animal models, AβOs have been shown to attach to synapses and instigate synapse dysfunction and deterioration. To establish the pathological mechanism of synaptic loss in AD, it will be important to identify the synaptic targets to which AβOs attach. Methods: An unbiased approach using far western ligand blots has identified three synaptic proteins to which AβOs specifically attach. These proteins (p100, p140, and p260) were subsequently enriched by detergent extraction, ultracentrifugation, and CHT-HPLC column separation, and sequenced by LC-MS/MS. P100, p140, and p260 were identified. These levels of AβOs targets in human AD and aging frontal cortexes were analyzed by quantitative proteomics and western-blot. The polyclonal antibody to AβOs was developed and used to block the toxicity of AβOs. The data were analyzed with one-way analysis of variance. Results: AβOs binding proteins p100, p140, and p260 were identified as Na/K-ATPase, synGap, and Shank3, respectively. α3-Na/K-ATPase, synGap, and Shank3 proteins showed loss in the postsynaptic density (PSD) of human AD frontal cortex. In short term experiments, oligomers of Aβ inhibited Na/K-ATPase at the synapse. Na/K-ATPase activity was restored by an antibody specific for soluble forms of Aβ. α3-Na/K-ATPase protein and synaptic β-amyloid peptides were pulled down from human AD synapses by co-immunoprecipitation. Results suggest synaptic dysfunction in early stages of AD may stem from inhibition of Na/K-ATPase activity by Aβ oligomers, while later stages could hypothetically result from disrupted synapse structure involving the PSD proteins synGap and Shank3. Conclusion: We identified three AβO binding proteins as α3-Na/K-ATPase, synGap, and Shank3. Soluble Aβ oligomers appear capable of attacking neurons via specific extracellular as well as intracellular synaptic proteins. Impact on these proteins hypothetically could lead to synaptic dysfunction and loss, and could serve as novel therapeutic targets for AD treatment by antibodies or other agents.
- Published
- 2019
- Full Text
- View/download PDF
28. IMMU-02. NEOANTIGENS ARISING FROM ALTERNATIVE SPLICING EVENTS MAY BE TARGETED BY TUMOR INFILTRATING LYMPHOCYTES IN GLIOBLASTOMAS
- Author
-
Frances Chow, Robert M. Prins, Anthony C. Wang, Linda M. Liau, Alexander Lee, Aaron Mochizuki, Richard Everson, Yi Xing, Mildred Galvez, David Nathanson, and Yang Pan
- Subjects
Cancer Research ,integumentary system ,Tumor-infiltrating lymphocytes ,medicine.medical_treatment ,Alternative splicing ,Immunology ,Cancer ,Immunotherapy ,Human leukocyte antigen ,Biology ,medicine.disease ,Immune system ,Oncology ,Antigen ,RNA splicing ,medicine ,Cancer research ,Neurology (clinical) - Abstract
INTRODUCTION Alternative splicing, the cellular process that converts premature mRNA to mature mRNA and allows for single genes to produce multiple protein products, is frequently dysregulated in many cancers, including glioblastoma. However, along with non-synonymous mutations in the DNA, altered splicing mechanisms in cancers may produce novel antigens (so-called neoantigens) that distinguish cancer cells from healthy cells and can thus be targeted by the immune system. METHODS We developed a new computation pipeline (IRIS – Isoform peptides from RNA splicing for Immunotherapy targets Screening) that took bulk RNA-sequencing data from 23 glioblastoma patient tumor samples and predicted neoantigens that may arise from alternative splicing events. We prioritized predicted neoantigens that arose in HLA*A02:01 and HLA*A03:01 patients and selected 8 potential neoantigens to generate peptide:MHC Class 1 dextramers. We tested PBMCs and/or ex vivo expanded tumor infiltrating lymphocytes (TIL) from 6 of our glioblastoma patients against these dextramers, sorted for any neoantigen-reactive T cells, and performed single-cell RNAsequencing on the sorted population to determine the TCR sequence. RESULTS Among the 8 predicted neoantigens tested, 7 of the neoantigens were recognized by at least 1 patient’s T cells. 1 HLA*A03:01 epitope was recognized in 3 of the 4 HLA*A03:01 patients tested and this epitope was highly positive in an expanded TIL population, representing 1.7% of all CD3+ CD8+ cells. When we sorted for those neoantigen reactive T cells from the expanded TIL population and performed single-cell RNAsequencing, we found 325 unique T cell clonotypes, but the top 10 clonotypes represented 83.6% of all TCR clonotypes. The most frequent TCR clonotype represented 39.1% of the repertoire and suggests that clonal expansion of a select few TCR clones occurred within the tumor. CONCLUSIONS In total, our data indicates that neoantigens arising from alternative splicing events may represent a potential target for immunotherapy in glioblastoma.
- Published
- 2019
29. TMIC-06. MYELOID POPULATIONS AND THE EFFECT OF NEOADJUVANT PD-1 INHIBITION IN THE GLIOBLASTOMA MICROENVIRONMENT: A SURFACEOMIC AND TRANSCRIPTOMIC DISSECTION AT THE SINGLE-CELL LEVEL
- Author
-
Robert M. Prins, Joey Orpilla, Mildred Galvez, Tom B. Davidson, Alexander Lee, Jenny Kienzler, Timothy F. Cloughesy, Linda M. Liau, Aaron Mochizuki, Anthony C. Wang, Frances Chow, and Richard Everson
- Subjects
Cancer Research ,Myeloid ,Cell cycle checkpoint ,medicine.medical_treatment ,Melanoma ,Pembrolizumab ,Immunotherapy ,Biology ,medicine.disease ,Transcriptome ,medicine.anatomical_structure ,Cytokine ,Oncology ,medicine ,Cancer research ,Tumor Microenvironment ,Neurology (clinical) ,Neoadjuvant therapy - Abstract
INTRODUCTION Glioblastoma (GBM) is the most common malignant brain tumor in adults and is associated with a dismal prognosis. Neoadjuvant anti-PD-1 blockade has demonstrated efficacy in melanoma, non-small cell lung cancer and recurrent GBM; however, responses vary. While T cells have garnered considerable attention in the context of immunotherapy, the role of myeloid cells in the GBM microenvironment remains controversial. METHODS We isolated CD45+ immune populations from patients who underwent brain tumor resection at UCLA. We hypothesized that myeloid cells in glioblastoma contribute to T cell dysfunction; however, this immune suppression can be mitigated by neoadjuvant PD-1 inhibition. To test this, we utilized mass cytometry and single-cell RNA sequencing to characterize these immune populations. RESULTS Mass cytometry profiling of tumor infiltrating lymphocytes from patients with GBM demonstrated a preponderance of CD11b+ myeloid populations (75% versus 25% CD3+). At the transcriptomic level, myeloid cells in newly diagnosed GBMs exhibited decreased expression of CCL4 (loge fold change -1.18, Bonferroni-adjusted P = 1.62x10-254) and its ligands compared to anaplastic astrocytoma. In ranked gene set enrichment analysis, patients who received neoadjuvant pembrolizumab demonstrated enrichment in TNFα-, NFκB- and lipid metabolism-related gene sets by bootstrapped Kolmogorov-Smirnov test (Benjamini-Hochberg adjusted P = 4.74x10-3, 1.45x10-2 and 2.48x10-3, respectively) in tumor-associated myeloid populations. Additionally, single-cell trajectory analysis demonstrated increased CCL4 and decreased ISG15 with neoadjuvant checkpoint inhibition. CONCLUSIONS Here, we utilize mass cytometry and single-cell RNA sequencing to demonstrate the predominance and transcriptomic features of myeloid populations in GBM. Myeloid cells in patients who receive neoadjuvant PD-1 blockade re-express increased levels NFκB, TNFα and CCL4, a cytokine crucial for the recruitment of dendritic cells to the tumor for antigen-specific T cell activation. By delving into the GBM microenvironment at the single-cell level, we hope to better delineate the role of myeloid populations in this uniformly fatal tumor.
- Published
- 2019
30. ATIM-16. VALIDATION OF RESPONSE TO NEOADJUVANT ANTI-PD-1 IMMUNOTHERAPY IN RECURRENT GLIOBLASTOMA
- Author
-
Timothy F. Cloughesy, Mildred Galvez, Robert M. Prins, Richard Everson, Frances Chow, Joey Orpilla, Alexander Lee, Linda M. Liau, and Aaron Mochizuki
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Adult Clinical Trials–Immunologic ,Recurrent glioblastoma ,medicine.medical_treatment ,Anti pd 1 ,Pembrolizumab ,Immunotherapy ,Tumor excision ,Internal medicine ,medicine ,Combined Modality Therapy ,Neurology (clinical) ,Progression-free survival ,business ,Neoadjuvant therapy - Abstract
BACKGROUND Recurrent glioblastoma has a poor median overall survival despite multimodal treatment. The use of pembrolizumab, an anti-PD-1 monoclonal antibody, demonstrates promise, including improved overall survival and increased immune response, when used in the neoadjuvant setting. METHODS We evaluated 13 patients with surgically accessible recurrent glioblastoma who were treated at the University of California, Los Angeles with neoadjuvant checkpoint inhibition prior to tumor resection. Combining data with our previously published multi-center study, we followed this cohort prospectively, performing tumor gene expression to validate the immune response and improved survival following neoadjuvant therapy with pembrolizumab. RESULTS When combined with findings from our prior study, patients in the neoadjuvant group (n = 27) had an overall survival of 400 days, whereas those in the adjuvant-only group (n = 19) had a median overall survival of 228 days by Kaplan-Meier estimator (P = 0.029, log-rank test). Progression-free survival in the neoadjuvant group was 108 days, whereas in the adjuvant-only group it was 72.5 days (P = 0.017, log-rank test). In elastic net penalized Cox proportional hazards regression, neoadjuvant anti-PD-1 blockade continued to be associated with improved overall survival, with a hazard ratio of 0.11 (P=0.003, log-rank test). Bulk tumor gene expression corroborates our previously described pattern of increased T-cell- and IFN-g-related gene expression, decreased cell-cycle-related signatures and their association with clinical response. Additionally, epigenetic regulation inversely correlated with clinical response. CONCLUSIONS Our findings support the neoadjuvant timing of PD-1 blockade in enhancing local immune responses and prolonging overall- and progression-free-survival. Correlative biological studies suggest a potential role for cell-cycle-related gene expression as a biomarker to predict response to therapy. Continued data collection and the addition of patients to the neoadjuvant arm of the trial, will be crucial to determine the potential role of checkpoint inhibition in the treatment of this deadly disease.
- Published
- 2019
- Full Text
- View/download PDF
31. Patterns of long-term survivorship following bevacizumab treatment for recurrent glioma: a case series
- Author
-
Albert Lai, Benjamin M. Ellingson, Frances Chow, Matthew Ji, Richard M. Green, Phioanh L. Nghiemphu, Linda M. Liau, Sean T Pianka, Donna Molaie, Liang Yen Liu, Nhung T Nguyen, and Timothy F. Cloughesy
- Subjects
Adult ,Male ,Oncology ,medicine.medical_specialty ,Long Term Survivorship ,recurrence ,Bevacizumab ,Angiogenesis Inhibitors ,bevacizumab ,Recurrent Glioma ,California ,Cohort Studies ,Young Adult ,Rare Diseases ,glioma ,Internal medicine ,Glioma ,medicine ,Humans ,Case Series ,Survivors ,Retrospective Studies ,Aged ,Cancer ,relapse ,Series (stratigraphy) ,business.industry ,glioblastoma ,Neurosciences ,General Medicine ,Middle Aged ,medicine.disease ,Brain Disorders ,Large cohort ,Brain Cancer ,Neoplasm Recurrence ,Treatment Outcome ,Local ,survivor ,Female ,long term ,lipids (amino acids, peptides, and proteins) ,progression ,Neoplasm Recurrence, Local ,business ,medicine.drug ,Glioblastoma - Abstract
Aim: Long-term survivors (LTS)after glioma recurrence while on bevacizumab (Bev)therapy are rarely reported in the current literature. The purpose of this case series is to confirm the existence of and describe a large cohort of recurrent glioma LTS treated with Bev (Bev-LTS). Patients & methods: We identified Bev-LTS as patients with post-Bev initiation survival times of ≥3 years among 1397 Bev treated recurrent glioma patients. Results: Among 962 grade-IV,221 grade III, and214 grade II Bev-treated glioma patients, we identified 28 (2.9%),14 (6.3%) and8(3.7%)Bev-LTS patients, respectively. 45Bev-LTS patients recurred on Bev, with 36 of those patients continuing therapy.Conclusion: Our study shows that a small portion of grade-IV, -III,and -II glioma patients can have long-term survival on Bev therapy even after Bev recurrence.
- Published
- 2019
- Full Text
- View/download PDF
32. Pathological Role of Peptidyl-Prolyl Isomerase Pin1 in the Disruption of Synaptic Plasticity in Alzheimer's Disease
- Author
-
Lingyan Xu, Richard Tsai, Carol F. Lippa, Flavio Rizzolio, Zhiyun Ren, Yungen Xu, Frances Chow, Yuesong Gong, Tongzheng Liu, Silvia Boffo, and Shaohui Huang
- Subjects
0301 basic medicine ,Dendritic spine ,Inbred C57BL ,Transgenic ,Mice ,Receptors ,80 and over ,Phosphorylation ,Cells, Cultured ,Aged, 80 and over ,Mice, Knockout ,Cultured ,Neuronal Plasticity ,Brain ,Long-term potentiation ,Cell biology ,Aged ,Alzheimer Disease ,Amyloid beta-Peptides ,Animals ,Dendritic Spines ,Disks Large Homolog 4 Protein ,Humans ,Mice, Inbred C57BL ,Mice, Transgenic ,NIMA-Interacting Peptidylprolyl Isomerase ,Nerve Tissue Proteins ,Post-Synaptic Density ,Receptors, N-Methyl-D-Aspartate ,Ubiquitin ,tau Proteins ,Neurology ,Neurology (clinical) ,NMDA receptor ,Alzheimer's disease ,N-Methyl-D-Aspartate ,Research Article ,Article Subject ,Cells ,Knockout ,Settore BIO/11 - Biologia Molecolare ,Biology ,lcsh:RC321-571 ,03 medical and health sciences ,medicine ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,medicine.disease ,030104 developmental biology ,Synaptic fatigue ,Proteasome ,Synaptic plasticity ,Postsynaptic density ,Neuroscience - Abstract
Synaptic loss is the structural basis for memory impairment in Alzheimer’s disease (AD). While the underlying pathological mechanism remains elusive, it is known that misfolded proteins accumulate as β-amyloid (Aβ) plaques and hyperphosphorylated Tau tangles decades before the onset of clinical disease. The loss of Pin1 facilitates the formation of these misfolded proteins in AD. Pin1 protein controls cell-cycle progression and determines the fate of proteins by the ubiquitin proteasome system. The activity of the ubiquitin proteasome system directly affects the functional and structural plasticity of the synapse. We localized Pin1 to dendritic rafts and postsynaptic density (PSD) and found the pathological loss of Pin1 within the synapses of AD brain cortical tissues. The loss of Pin1 activity may alter the ubiquitin-regulated modification of PSD proteins and decrease levels of Shank protein, resulting in aberrant synaptic structure. The loss of Pin1 activity, induced by oxidative stress, may also render neurons more susceptible to the toxicity of oligomers of Aβ and to excitation, thereby inhibiting NMDA receptor-mediated synaptic plasticity and exacerbating NMDA receptor-mediated synaptic degeneration. These results suggest that loss of Pin1 activity could lead to the loss of synaptic plasticity in the development of AD.
- Published
- 2016
33. Temozolomide and Other Potential Agents for the Treatment of Glioblastoma Multiforme
- Author
-
Daniel T. Nagasawa, Frances Chow, Andrew Yew, Isaac Yang, Won Kim, and Nicole Cremer
- Subjects
Oncology ,medicine.medical_specialty ,Standard of care ,Bevacizumab ,Internal medicine ,Temozolomide ,Humans ,Medicine ,Treatment resistance ,Antineoplastic Agents, Alkylating ,Survival rate ,Brain Neoplasms ,business.industry ,General Medicine ,medicine.disease ,Dacarbazine ,Treatment Outcome ,Surgery ,Neurology (clinical) ,Glioblastoma ,business ,Stage iv ,Median survival ,medicine.drug - Abstract
This article provides historical and recent perspectives related to the use of temozolomide for the treatment of glioblastoma multiforme. Temozolomide has quickly become part of the standard of care for the modern treatment of stage IV glioblastoma multiforme since its approval in 2005. Yet despite its improvements from previous therapies, median survival remains approximately 15 months, with a 2-year survival rate of 8% to 26%. The mechanism of action of this chemotherapeutic agent, conferred advantages and limitations, treatment resistance and rescue, and potential targets of future research are discussed.
- Published
- 2012
- Full Text
- View/download PDF
34. The Challenge of Distinguishing POEMS from Chronic Inflammatory Demyelinating Polyneuropathy—Importance of Early Recognition and Diagnosis of POEMS
- Author
-
Said R. Beydoun, Leila Darki, and Frances Chow
- Subjects
03 medical and health sciences ,medicine.medical_specialty ,0302 clinical medicine ,business.industry ,030220 oncology & carcinogenesis ,General Neuroscience ,Medicine ,Chronic inflammatory demyelinating polyneuropathy ,Neurology (clinical) ,business ,medicine.disease ,Dermatology ,030217 neurology & neurosurgery - Abstract
POEMS is a rare syndrome characterized by the unique constellation of polyneuropathy, organomegaly, endocrinopathy, M-proteins, and skin changes. Correct diagnosis is often delayed in early stages of the syndrome when patients exhibit only isolated polyneuropathy due to the clinical and electrodiagnostic similarities with chronic inflammatory demyelinating polyneuropathy. We describe a case in which early suspicion for POEMS uncovered underlying malignancy, and we review the clinical, electrophysiological, pathological, and laboratory findings characteristic of POEMS. The importance of high clinical suspicion is key in the proper diagnosis and management of this complex syndrome.
- Published
- 2018
- Full Text
- View/download PDF
35. The Potential Role of Insulin on the Shank-Postsynaptic Platform in Neurodegenerative Diseases Involving Cognition
- Author
-
Yuesong Gong, Frances Chow, and Carol F. Lippa
- Subjects
medicine.medical_treatment ,Central nervous system ,Nerve Tissue Proteins ,Disease ,Pathogenesis ,Postsynaptic potential ,mental disorders ,Medicine ,Animals ,Humans ,Insulin ,business.industry ,General Neuroscience ,Post-Synaptic Density ,Cognition ,Neurodegenerative Diseases ,medicine.disease ,body regions ,Psychiatry and Mental health ,Clinical Psychology ,Synaptic function ,medicine.anatomical_structure ,Schizophrenia ,Geriatrics and Gerontology ,business ,Neuroscience - Abstract
Loss of synaptic function is critical in the pathogenesis of Alzheimer’s disease (AD) and other central nervous system (CNS) degenerations. A promising candidate in the regulation of synaptic function is Shank, a protein that serves as a scaffold for excitatory synaptic receptors and proteins. Loss of Shank alters structure and function of the postsynaptic density (PSD). Shank proteins are associated with N-methyl-d-aspartate and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor loss at the PSD in AD; mutations in Shank also lead to autism spectrum disorders (ASDs) and schizophrenia, both of which affect cognition, suggesting that Shank may play a common pathologic role in AD, ASD, and schizophrenia. Shank protein directly associates with insulin receptor substrate protein p53 in PSD. Insulin and insulin sensitizers have been used in clinical trials for these diseases; this suggests that insulin signals may alter protein homeostasis at the shank-postsynaptic platform in PSDs; insulin could improve the function of synapses in these diseases.
- Published
- 2014
36. P4–020: Pathogenic role of Pin1 in synaptic dysfunction in Alzheimer's disease
- Author
-
Frances Chow, Yuesong Gong, and Carol F. Lippa
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,business.industry ,Health Policy ,PIN1 ,Medicine ,Neurology (clinical) ,Disease ,Geriatrics and Gerontology ,business ,Neuroscience - Published
- 2013
- Full Text
- View/download PDF
37. Molecular characteristics and pathways of Avastin for the treatment of glioblastoma multiforme
- Author
-
Marko Spasic, Frances Chow, Isaac Yang, Daniel T. Nagasawa, and Claire Tu
- Subjects
Oncology ,Vascular Endothelial Growth Factor A ,medicine.medical_specialty ,genetic structures ,Bevacizumab ,medicine.medical_treatment ,Angiogenesis Inhibitors ,Antibodies, Monoclonal, Humanized ,Disease-Free Survival ,Food and drug administration ,Internal medicine ,medicine ,Humans ,Adverse effect ,Chemotherapy ,business.industry ,Brain Neoplasms ,General Medicine ,medicine.disease ,Primary tumor ,Gene Expression Regulation, Neoplastic ,Invasive phenotype ,Monoclonal ,Surgery ,Neurology (clinical) ,Neoplasm Recurrence, Local ,business ,Glioblastoma ,medicine.drug - Abstract
This article provides historical background and current research involving the use of bevacizumab for the treatment of recurrent glioblastoma. Although bevacizumab, approved by the Food and Drug Administration, prolongs glioblastoma progression free survivial, decreases tumor vascularization, and reduces permeability of vessels, it does not seem to prolong overall survival. Despite slowed primary tumor progression, bevacizumab treatment may facilitate transformation to a more invasive phenotype. Adaptive responses, which make glioblastoma particularly resistant to various treatment modalities have been described. Conferred benefits, adverse effects, mechanisms of resistance, and potential areas for future research are discussed.
- Published
- 2012
38. Peptidyl-Prolyl Isomerase Pin1: A Common Therapeutic Target for Synaptic Dysfunction and Protein Misfolding in Alzheimer's Disease (SC02.008)
- Author
-
Richard Tsai, Frances Chow, Yuesong Gong, and Carol F. Lippa
- Subjects
Synapse ,Dendritic spine ,Excitatory synapse ,Biochemistry ,Postsynaptic potential ,Immunocytochemistry ,Prolyl isomerase ,PIN1 ,NMDA receptor ,Neurology (clinical) ,Biology ,Cell biology - Abstract
Objective: The Pin1 protein, a peptidyl-prolyl isomerase, facilitates proper folding of proteins. Loss of Pin1 in mice induces formation of misfolded beta-amyloid and hyperphosphorylated tau at dendritic rafts, which suggests that Pin1 may pathologically alter dendritic rafts and the post-synaptic density (PSD) in Alzheimer9s disease (AD). In this study, we observed Pin1 associations with synaptic proteins and compared Pin1 pathological changes in AD patients against controls. Background The structural basis for memory formation lies in the neuronal excitatory synapse. Unique to this synapse and underlying its function are integrated detergent-resistant dendritic rafts and PSD within the postsynaptic structure. The association of the pathological hallmarks of AD--misfolded oligomers of beta-amyloid and hyperphosphorylated tau--with these detergent-resistant microdomains may lead to synaptic dysfunction and loss. Design/Methods: Human cortices from 12 AD patients and 10 age-matched controls were prepared using a tissue extraction protocol, followed by SDS-PAGE, Western and Dot blot analyses. Mice cortical neurons were cultured and visualized using immunocytochemistry, immunoblot, and confocal microscopy. Results: We found that in AD, Pin1 proteins coincidently associated with NMDA receptor 1, Shank3 proteins, and misfolded proteins at dendritic rafts and the PSD. The protein levels of Pin1 were pathologically increased in dendritic rafts and decreased in the PSD of AD brains. In cultured neurons, Pin1 proteins co-localized with NMDA receptor 1 and Shank3, and dendritic spines degenerated after Pin1 activity was blocked. Conclusions: Our findings suggest that pathological distribution and loss of Pin1 at the synapse may have a direct effect on phosphorylation and homeostasis of proteins beside beta-amyloid and hyperphosphorylated tau in AD development. Pin1 may prove to be a common therapeutic target for both the treatment of synaptic dysfunction and misfolding of beta-amyloid and phosphorylated tau at synapses in early stages of AD. Disclosure: Dr. Chow has nothing to disclose. Dr. Tsai has nothing to disclose. Dr. Lippa has received personal compensation in an editorial capacity for Sage Publications.Dr. Lippa has received research support from UCB Pharma and DUCOM. Dr. Gong has nothing to disclose.
- Published
- 2012
- Full Text
- View/download PDF
39. Phosphoenol pyruvamides. Amide-phosphate interactions in analogs of phosphoenol pyruvate
- Author
-
Jane Frances Chow, Ronald Kluger, and James J. Croke
- Subjects
chemistry.chemical_compound ,Colloid and Surface Chemistry ,Chemistry ,Stereochemistry ,Amide ,General Chemistry ,Phosphate ,Biochemistry ,Catalysis - Abstract
Hydrolyse du propylamide et du phenylamide de l'acide diethoxyphosphoryl-2 acrylique. Constantes de vitesse. Mecanisme. Effet des substituants
- Published
- 1984
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.