324 results on '"Ashley, David M."'
Search Results
302. Pooled genetic screens to identify vulnerabilities in TERT-promoter-mutant glioblastoma.
- Author
-
Tu KJ, Stewart CE, Hendrickson PG, Regal JA, Kim SY, Ashley DM, Waitkus MS, and Reitman ZJ
- Subjects
- Humans, Gene Regulatory Networks, Promoter Regions, Genetic genetics, Mutation, Transcription Factors genetics, Cell Line, Tumor, Glioblastoma genetics, Telomerase genetics
- Abstract
Pooled genetic screens represent a powerful approach to identify vulnerabilities in cancer. Here we used pooled CRISPR/Cas9-based approaches to identify vulnerabilities associated with telomerase reverse transcriptase (TERT) promoter mutations (TPMs) found in >80% of glioblastomas. We first developed a platform to detect perturbations that cause long-term growth defects in a TPM-mutated glioblastoma cell line. However, we could not detect dependencies on either TERT itself or on an E-twenty six transcription (ETS) factor known to activate TPMs. To explore this finding, we cataloged TPM status for 441 cell lines and correlated this with genome-wide screening data. We found that TPM status was not associated with differential dependency on TERT, but that E-twenty six (ETS) transcription factors represent key dependencies in both TPM+ and TPM- lines. Further, we found that TPMs are associated with expression of gene programs regulated by a wide array of ETS-factors in both cell lines and primary glioblastoma tissues. This work contributes a unique TPM cell line reagent, establishes TPM status for many deeply-profiled cell lines, and catalogs TPM-associated vulnerabilities. The results highlight challenges in executing genetic screens to detect TPM-specific vulnerabilities, and suggest redundancy in the genetic network that regulates TPM function with therapeutic implications., (© 2023. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
303. NUTMEG: A randomized phase II study of nivolumab and temozolomide versus temozolomide alone in newly diagnosed older patients with glioblastoma.
- Author
-
Sim HW, Wachsmuth L, Barnes EH, Yip S, Koh ES, Hall M, Jennens R, Ashley DM, Verhaak RG, Heimberger AB, Rosenthal MA, Hovey EJ, Ellingson BM, Tognela A, Gan HK, Wheeler H, Back M, McDonald KL, Long A, Cuff K, Begbie S, Gedye C, Mislang A, Le H, Johnson MO, Kong BY, Simes JR, Lwin Z, and Khasraw M
- Abstract
Background: There is an immunologic rationale to evaluate immunotherapy in the older glioblastoma population, who have been underrepresented in prior trials. The NUTMEG study evaluated the combination of nivolumab and temozolomide in patients with glioblastoma aged 65 years and older., Methods: NUTMEG was a multicenter 2:1 randomized phase II trial for patients with newly diagnosed glioblastoma aged 65 years and older. The experimental arm consisted of hypofractionated chemoradiation with temozolomide, then adjuvant nivolumab and temozolomide. The standard arm consisted of hypofractionated chemoradiation with temozolomide, then adjuvant temozolomide. The primary objective was to improve overall survival (OS) in the experimental arm., Results: A total of 103 participants were randomized, with 69 in the experimental arm and 34 in the standard arm. The median (range) age was 73 (65-88) years. After 37 months of follow-up, the median OS was 11.6 months (95% CI, 9.7-13.4) in the experimental arm and 11.8 months (95% CI, 8.3-14.8) in the standard arm. For the experimental arm relative to the standard arm, the OS hazard ratio was 0.85 (95% CI, 0.54-1.33). In the experimental arm, there were three grade 3 immune-related adverse events which resolved, with no unexpected serious adverse events., Conclusions: Due to insufficient evidence of benefit with nivolumab, the decision was made not to transition to a phase III trial. No new safety signals were identified with nivolumab. This complements the existing series of immunotherapy trials. Research is needed to identify biomarkers and new strategies including combinations., Competing Interests: HWS acknowledges institutional research funding from AbbVie and Bristol-Myers Squibb and has received honoraria from Eli Lilly. ABH serves on the advisory board of Caris Life Sciences and WIRB-Copernicus Group, and is supported by research grants from AbbVie, Alnylam Pharmaceuticals and Cellularity. BME serves on the advisory board and as a consultant for Alpheus Medical, Curtana Pharmaceuticals, Ellipses Pharma, Global Coalition for Adaptive Research, ImmunoGenesis, Medicenna, MedQIA, Monteris, Neosoma, Sagimet Biosciences, Sapience Therapeutics, Servier Pharmaceuticals, Siemens and Sumitomo Pharma Oncology, serves as a consultant for Carthera and Chimerix, and is supported by a research grant from Siemens. HG serves on the advisory board of Merck Serono, and serves as a consultant for Curis and Telix Pharmaceuticals. HL acknowledges institutional research funding from Pfizer, and has received honoraria from AstraZeneca. JRS acknowledges institutional research funding from AbbVie, Amgen, Astellas, AstraZeneca, Bayer, Bristol-Myers Squibb, Cancer Australia, Cancer Institute NSW, Merck Serono, National Health and Medical Research Council of Australia, Pfizer and Roche. ZL serves on the scientific advisory committee of Merck Sharp Dohme, has received honoraria from AstraZeneca, Bristol-Myers Squibb and Roche, and has received travel funding from Pfizer. MK acknowledges institutional research funding from AbbVie, Bristol-Myers Squibb and Specialized Therapeutics, and has received honoraria from AbbVie, Bristol-Myers Squibb, Eli Lilly, Ipsen, Pfizer and Roche., (© 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
304. Polio virotherapy targets the malignant glioma myeloid infiltrate with diffuse microglia activation engulfing the CNS.
- Author
-
Yang Y, Brown MC, Zhang G, Stevenson K, Mohme M, Kornahrens R, Bigner DD, Ashley DM, López GY, and Gromeier M
- Subjects
- Animals, Mice, Microglia metabolism, B7-H1 Antigen, Inflammation, Oncolytic Virotherapy, Glioma therapy, Brain Neoplasms metabolism, Poliomyelitis therapy
- Abstract
Background: Malignant gliomas commandeer dense inflammatory infiltrates with glioma-associated macrophages and microglia (GAMM) promoting immune suppression, evasion, and tumor progression. Like all cells in the mononuclear phagocytic system, GAMM constitutively express the poliovirus receptor, CD155. Besides myeloid cells, CD155 is widely upregulated in the neoplastic compartment of malignant gliomas. Intratumor treatment with the highly attenuated rhino:poliovirus chimera, PVSRIPO, yielded long-term survival with durable radiographic responses in patients with recurrent glioblastoma (Desjardins et al. New England Journal of Medicine, 2018). This scenario raises questions about the contributions of myeloid versus neoplastic cells to polio virotherapy of malignant gliomas., Methods: We investigated PVSRIPO immunotherapy in immunocompetent mouse brain tumor models with blinded, board-certified neuropathologist review, a range of neuropathological, immunohistochemical, and immunofluorescence analyses, and RNAseq of the tumor region., Results: PVSRIPO treatment caused intense engagement of the GAMM infiltrate associated with substantial, but transient tumor regression. This was accompanied by marked microglia activation and proliferation in normal brain surrounding the tumor, in the ipsilateral hemisphere and extending into the contralateral hemisphere. There was no evidence for lytic infection of malignant cells. PVSRIPO-instigated microglia activation occurred against a backdrop of sustained innate antiviral inflammation, associated with induction of the Programmed Cell Death Ligand 1 (PD-L1) immune checkpoint on GAMM. Combining PVSRIPO with PD1/PD-L1 blockade led to durable remissions., Conclusions: Our work implicates GAMM as active drivers of PVSRIPO-induced antitumor inflammation and reveals profound and widespread neuroinflammatory activation of the brain-resident myeloid compartment by PVSRIPO., (© The Author(s) 2023. 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
- 2023
- Full Text
- View/download PDF
305. Cancer-associated SMARCAL1 loss-of-function mutations promote alternative lengthening of telomeres and tumorigenesis in telomerase-negative glioblastoma cells.
- Author
-
Liu H, Xu C, Diplas BH, Brown A, Strickland LM, Yao H, Ling J, McLendon RE, Keir ST, Ashley DM, He Y, and Waitkus MS
- Subjects
- Humans, Telomere Homeostasis, Mutation, Telomere genetics, Telomere metabolism, Carcinogenesis, Cell Transformation, Neoplastic genetics, DNA Helicases genetics, DNA Helicases metabolism, Telomerase genetics, Telomerase metabolism, Glioblastoma genetics
- Abstract
Background: Telomere maintenance mechanisms are required to enable the replicative immortality of malignant cells. While most cancers activate the enzyme telomerase, a subset of cancers uses telomerase-independent mechanisms termed alternative lengthening of telomeres (ALT). ALT occurs via homology-directed-repair mechanisms and is frequently associated with ATRX mutations. We previously showed that a subset of adult glioblastoma (GBM) patients with ATRX-expressing ALT-positive tumors harbored loss-of-function mutations in the SMARCAL1 gene, which encodes an annealing helicase involved in replication fork remodeling and the resolution of replication stress. However, the causative relationship between SMARCAL1 deficiency, tumorigenesis, and de novo telomere synthesis is not understood., Methods: We used a patient-derived ALT-positive GBM cell line with native SMARCAL1 deficiency to investigate the role of SMARCAL1 in ALT-mediated de novo telomere synthesis, replication stress, and gliomagenesis in vivo., Results: Inducible rescue of SMARCAL1 expression suppresses ALT indicators and inhibits de novo telomere synthesis in GBM and osteosarcoma cells, suggesting that SMARCAL1 deficiency plays a functional role in ALT induction in cancers that natively lack SMARCAL1 function. SMARCAL1-deficient ALT-positive cells can be serially propagated in vivo in the absence of detectable telomerase activity, demonstrating that the SMARCAL1-deficient ALT phenotype maintains telomeres in a manner that promotes tumorigenesis., Conclusions: SMARCAL1 deficiency is permissive to ALT and promotes gliomagenesis. Inducible rescue of SMARCAL1 in ALT-positive cell lines permits the dynamic modulation of ALT activity, which will be valuable for future studies aimed at understanding the mechanisms of ALT and identifying novel anticancer therapeutics that target the ALT phenotype., (© The Author(s) 2023. 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
- 2023
- Full Text
- View/download PDF
306. Recombinant polio-rhinovirus immunotherapy for recurrent paediatric high-grade glioma: a phase 1b trial.
- Author
-
Thompson EM, Landi D, Brown MC, Friedman HS, McLendon R, Herndon JE 2nd, Buckley E, Bolognesi DP, Lipp E, Schroeder K, Becher OJ, Friedman AH, McKay Z, Walter A, Threatt S, Jaggers D, Desjardins A, Gromeier M, Bigner DD, and Ashley DM
- Subjects
- Adult, Humans, Child, Male, Female, Adolescent, Rhinovirus, Neoplasm Recurrence, Local therapy, Immunotherapy, Glioblastoma, Glioma drug therapy, Brain Neoplasms therapy, Astrocytoma, Poliomyelitis, Cerebellar Neoplasms
- Abstract
Background: Outcomes of recurrent paediatric high-grade glioma are poor, with a median overall survival of less than 6 months. Viral immunotherapy, such as the polio-rhinovirus chimera lerapolturev, is a novel approach for treatment of recurrent paediatric high-grade glioma and has shown promise in adults with recurrent glioblastoma. The poliovirus receptor CD155 is ubiquitously expressed in malignant paediatric brain tumours and is a treatment target in paediatric high-grade glioma. We aimed to assess the safety of lerapolturev when administered as a single dose intracerebrally by convection enhanced delivery in children and young people with recurrent WHO grade 3 or grade 4 glioma, and to assess overall survival in these patients., Methods: This phase 1b trial was done at the Duke University Medical Center (Durham, NC, USA). Patients aged 4-21 years with recurrent high-grade malignant glioma (anaplastic astrocytoma, glioblastoma, anaplastic oligoastrocytoma, anaplastic oligodendroglioma, or anaplastic pleomorphic xanthoastrocytoma) or anaplastic ependymoma, atypical teratoid rhabdoid tumour, or medulloblastoma with infusible disease were eligible for this study. A catheter was tunnelled beneath the scalp for a distance of at least 5 cm to aid in prevention of infection. The next day, lerapolturev at a dose of 5 × 10
7 median tissue culture infectious dose in 3 mL infusate loaded in a syringe was administered via a pump at a rate of 0·5 mL per h as a one-time dose. The infusion time was approximately 6·5 h to compensate for volume of the tubing. The primary endpoint was the proportion of patients with unacceptable toxic effects during the 14-day period after lerapolturev treatment. The study is registered with ClinicalTrials.gov, NCT03043391., Findings: Between Dec 5, 2017, and May 12, 2021, 12 patients (11 unique patients) were enrolled in the trial. Eight patients were treated with lerapolturev. The median patient age was 16·5 years (IQR 11·0-18·0), five (63%) of eight patients were male and three (38%) were female, and six (75%) of eight patients were White and two (25%) were Black or African American. The median number of previous chemotherapeutic regimens was 3·50 (IQR 1·25-5·00). Six of eight patients had 26 treatment-related adverse events attributable to lerapolturev. There were no irreversible (ie, persisted longer than 2 weeks) treatment-related grade 4 adverse events or deaths. Treatment-related grade 3 adverse events included headaches in two patients and seizure in one patient. Four patients received low-dose bevacizumab on-study for treatment-related peritumoural inflammation or oedema, diagnosed by both clinical symptoms plus fluid-attenuated inversion recovery MRI. The median overall survival was 4·1 months (95% CI 1·2-10·1). One patient remains alive after 22 months., Interpretation: Convection enhanced delivery of lerapolturev is safe enough in the treatment of recurrent paediatric high-grade glioma to proceed to the next phase of trial., Funding: Solving Kids Cancer, B+ Foundation, Musella Foundation, and National Institutes of Health., Competing Interests: Declaration of interests MG, HSF, DPB, AD, DDB, and DMA are equity shareholders of Istari Oncology, the company licensing lerapolturev. MCB, MG, DDB, and DMA own intellectual property licensed to Istari Oncology. MCB, MG, DDB, HSF, and AD received consultancy fees from Istari Oncology. EMT is a scientific consultant for Oncoheroes Biosciences. All other authors declare no competing interests., (Copyright © 2023 Elsevier Ltd. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
307. Use of Trametinib in Children and Young Adults With Progressive Low-grade Glioma and Glioneuronal Tumors.
- Author
-
Hanzlik E, Archambault B, El-Dairi M, Schroeder K, Patel MP, Lipp ES, Peters KB, Ashley DM, and Landi D
- Subjects
- Child, Humans, Young Adult, Retrospective Studies, Pyridones therapeutic use, Brain Neoplasms pathology, Glioma pathology, Central Nervous System Neoplasms drug therapy
- Abstract
Low-grade gliomas/glioneuronal tumors comprise one-third of all pediatric-type CNS tumors. These tumors are generally caused by activating mutations in the mitogen-activated protein kinase (MAPK) pathway. Targeted drugs, such as trametinib, have shown promise in other cancers and are being utilized in low-grade gliomas. A retrospective chart review was conducted to evaluate radiographic response, visual outcomes, tolerability, and durability of response in progressive circumscribed low-grade gliomas treated with trametinib. Eleven patients were treated with trametinib. The best radiographic response was 2/11 partial response, 3/11 minor response, 3/11 stable disease, and 3/13 progressive disease. In the patients with partial or minor response, the best response was seen after longer durations of therapy; 4 of 5 best responses occurred after at least 9 months of therapy with a median of 21 months. Patients with optic pathway tumors showed at least stable vision throughout treatment, with 3 having improved vision on treatment. Trametinib is effective and well-tolerated in patients with progressive low-grade glioma. Best responses were seen after a longer duration of therapy in those with a positive response. Patients with optic pathway lesions showed stable to improved vision while on treatment., Competing Interests: K.B.P. has research support for unrelated projects from Servier, Biomimitex, Sapience, Varian, and Abbvie. KBP is a member of the advisory boards of Vivicitas Oncology and Cordance. The remaining authors declare no conflicts of interest., (Copyright © 2022 Wolters Kluwer Health, Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
308. The Enduring Effects of COVID for Cancer Care: Learning from Real-Life Clinical Practice.
- Author
-
Broom A, Williams Veazey L, Kenny K, Harper I, Peterie M, Page A, Cort N, Durling J, Lipp ES, Tan AC, Walsh KM, Hanks BA, Johnson M, Van Swearingen AED, Anders CK, Ashley DM, and Khasraw M
- Subjects
- Humans, Australia epidemiology, Pandemics, COVID-19 epidemiology, Neoplasms epidemiology, Neoplasms therapy
- Abstract
For three years, COVID-19 has circulated among our communities and around the world, fundamentally changing social interactions, health care systems, and service delivery. For people living with (and receiving treatment for) cancer, pandemic conditions presented significant additional hurdles in an already unstable and shifting environment, including disrupted personal contact with care providers, interrupted access to clinical trials, distanced therapeutic encounters, multiple immune vulnerabilities, and new forms of financial precarity. In a 2020 perspective in this journal, we examined how COVID-19 was reshaping cancer care in the early stages of the pandemic and how these changes might endure into the future. Three years later, and in light of a series of interviews with patients and their caregivers from the United States and Australia conducted during the pandemic, we return to consider the potential legacy effects of the pandemic on cancer care. While some challenges to care provision and survivorship were unforeseen, others accentuated and amplified existing problems experienced by patients, caregivers, and health care providers. Both are likely to have enduring effects in the "post-pandemic" world, raising the importance of focusing on lessons that can be learned for the future., (©2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
309. Interplay between ATRX and IDH1 mutations governs innate immune responses in diffuse gliomas.
- Author
-
Hariharan S, Whitfield BT, Pirozzi CJ, Waitkus MS, Brown MC, Bowie ML, Irvin DM, Roso K, Fuller R, Hostettler J, Dharmaiah S, Gibson EA, Briley A, Mangoli A, Fraley C, Shobande M, Stevenson K, Zhang G, Malgulwar PB, Roberts H, Roskoski M, Spasojevic I, Keir ST, He Y, Castro MG, Huse JT, and Ashley DM
- Abstract
Stimulating the innate immune system has been explored as a therapeutic option for the treatment of gliomas. Inactivating mutations in ATRX , defining molecular alterations in IDH -mutant astrocytomas, have been implicated in dysfunctional immune signaling. However, little is known about the interplay between ATRX loss and IDH mutation on innate immunity. To explore this, we generated ATRX knockout glioma models in the presence and absence of the IDH1
R 132 H mutation. ATRX-deficient glioma cells were sensitive to dsRNA-based innate immune agonism and exhibited impaired lethality and increased T-cell infiltration in vivo . However, the presence of IDH1R 132 H dampened baseline expression of key innate immune genes and cytokines in a manner restored by genetic and pharmacological IDH1R132H inhibition. IDH1R132H co-expression did not interfere with the ATRX KO-mediated sensitivity to dsRNA. Thus, ATRX loss primes cells for recognition of dsRNA, while IDH1R132H reversibly masks this priming. This work reveals innate immunity as a therapeutic vulnerability of astrocytoma.- Published
- 2023
- Full Text
- View/download PDF
310. Intratumor childhood vaccine-specific CD4 + T-cell recall coordinates antitumor CD8 + T cells and eosinophils.
- Author
-
Brown MC, Beasley GM, McKay ZP, Yang Y, Desjardins A, Randazzo DM, Landi D, Ashley DM, Bigner DD, Nair SK, and Gromeier M
- Subjects
- Mice, Humans, Animals, CD8-Positive T-Lymphocytes, Eosinophils, CD40 Ligand, Immunity, Innate, Lymphocytes, T-Lymphocytes, Regulatory, Tetanus, Vaccines
- Abstract
Background: Antitumor mechanisms of CD4
+ T cells remain crudely defined, and means to effectively harness CD4+ T-cell help for cancer immunotherapy are lacking. Pre-existing memory CD4+ T cells hold potential to be leveraged for this purpose. Moreover, the role of pre-existing immunity in virotherapy, particularly recombinant poliovirus immunotherapy where childhood polio vaccine specific immunity is ubiquitous, remains unclear. Here we tested the hypothesis that childhood vaccine-specific memory T cells mediate antitumor immunotherapy and contribute to the antitumor efficacy of polio virotherapy., Methods: The impact of polio immunization on polio virotherapy, and the antitumor effects of polio and tetanus recall were tested in syngeneic murine melanoma and breast cancer models. CD8+ T-cell and B-cell knockout, CD4+ T-cell depletion, CD4+ T-cell adoptive transfer, CD40L blockade, assessments of antitumor T-cell immunity, and eosinophil depletion defined antitumor mechanisms of recall antigens. Pan-cancer transcriptome data sets and polio virotherapy clinical trial correlates were used to assess the relevance of these findings in humans., Results: Prior vaccination against poliovirus substantially bolstered the antitumor efficacy of polio virotherapy in mice, and intratumor recall of poliovirus or tetanus immunity delayed tumor growth. Intratumor recall antigens augmented antitumor T-cell function, caused marked tumor infiltration of type 2 innate lymphoid cells and eosinophils, and decreased proportions of regulatory T cells (Tregs). Antitumor effects of recall antigens were mediated by CD4+ T cells, limited by B cells, independent of CD40L, and dependent on eosinophils and CD8+ T cells. An inverse relationship between eosinophil and Treg signatures was observed across The Cancer Genome Atlas (TCGA) cancer types, and eosinophil depletion prevented Treg reductions after polio recall. Pretreatment polio neutralizing antibody titers were higher in patients living longer, and eosinophil levels increased in the majority of patients, after polio virotherapy., Conclusion: Pre-existing anti-polio immunity contributes to the antitumor efficacy of polio virotherapy. This work defines cancer immunotherapy potential of childhood vaccines, reveals their utility to engage CD4+ T-cell help for antitumor CD8+ T cells, and implicates eosinophils as antitumor effectors of CD4+ T cells., Competing Interests: Competing interests: MCB, AD, DMA, DDB, SKN, and MG own intellectual property related to Lerapolturev, which has been licensed to Istari Oncology. MCB, AD, MG, and DDB received consultancy fees from Istari Oncology; MG and DDB hold equity in Istari Oncology. All other authors declare they have no competing interests., (© Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2023
- Full Text
- View/download PDF
311. Neuronal CaMKK2 promotes immunosuppression and checkpoint blockade resistance in glioblastoma.
- Author
-
Tomaszewski WH, Waibl-Polania J, Chakraborty M, Perera J, Ratiu J, Miggelbrink A, McDonnell DP, Khasraw M, Ashley DM, Fecci PE, Racioppi L, Sanchez-Perez L, Gunn MD, and Sampson JH
- Subjects
- Humans, CD8-Positive T-Lymphocytes, Tumor Microenvironment, Immunosuppression Therapy, Neurons pathology, Calcium-Calmodulin-Dependent Protein Kinase Kinase genetics, Glioblastoma drug therapy, Glioblastoma genetics
- Abstract
Glioblastoma (GBM) is notorious for its immunosuppressive tumor microenvironment (TME) and is refractory to immune checkpoint blockade (ICB). Here, we identify calmodulin-dependent kinase kinase 2 (CaMKK2) as a driver of ICB resistance. CaMKK2 is highly expressed in pro-tumor cells and is associated with worsened survival in patients with GBM. Host CaMKK2, specifically, reduces survival and promotes ICB resistance. Multimodal profiling of the TME reveals that CaMKK2 is associated with several ICB resistance-associated immune phenotypes. CaMKK2 promotes exhaustion in CD8
+ T cells and reduces the expansion of effector CD4+ T cells, additionally limiting their tumor penetrance. CaMKK2 also maintains myeloid cells in a disease-associated microglia-like phenotype. Lastly, neuronal CaMKK2 is required for maintaining the ICB resistance-associated myeloid phenotype, is deleterious to survival, and promotes ICB resistance. Our findings reveal CaMKK2 as a contributor to ICB resistance and identify neurons as a driver of immunotherapeutic resistance in GBM., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
312. cGAS-STING pathway targeted therapies and their applications in the treatment of high-grade glioma.
- Author
-
Tripathi S, Najem H, Mahajan AS, Zhang P, Low JT, Stegh AH, Curran MA, Ashley DM, James CD, and Heimberger AB
- Subjects
- Humans, Interferons, Nucleotidyltransferases genetics, Nucleotidyltransferases metabolism, Tumor Microenvironment, Glioma drug therapy, Membrane Proteins metabolism
- Abstract
Median survival of patients with glioblastoma (GBM) treated with standard of care which consists of maximal safe resection of the contrast-enhancing portion of the tumor followed by radiation therapy with concomitant adjuvant temozolomide (TMZ) remains 15 months. The tumor microenvironment (TME) is known to contain immune suppressive myeloid cells with minimal effector T cell infiltration. Stimulator of interferon genes (STING) is an important activator of immune response and results in production of Type 1 interferon and antigen presentation by myeloid cells. This review will discuss important developments in STING agonists, potential biomarkers for STING response, and new combinatorial therapeutic approaches in gliomas., Competing Interests: Competing interests: ABH has licensed intellectual property that has generated royalty income from Celldex Therapeutics and DNAtrix, serves on the advisory boards of WCG Oncology and Caris Life Sciences, receives research support from AbbVie, Moleculin, Celularity, Codiak Biosciences, and received consulting fees from Novocure and Istari Oncology. A.H.S. is shareholder of Exicure Inc., which develops the Spherical Nucleic Acid (SNA) therapeutic platforms. A.H.S. is inventor on patent US20150031745A1, which describes SNA nanoconjugates to cross the BBB, and on provisional patent 30938/2021-221P, describing SNAs to target the cGAS-STING pathway. DMA serves on the advisory boards of Diverse Bio Pharma, Immunogenesis, MAIA Biotech Inc., Jackson Laboratory. The rest of the authors declare no potential conflict of interest., (Copyright: © 2022 Tripathi S et al.)
- Published
- 2022
- Full Text
- View/download PDF
313. Antitumor Activity of a Mitochondrial-Targeted HSP90 Inhibitor in Gliomas.
- Author
-
Wei S, Yin D, Yu S, Lin X, Savani MR, Du K, Ku Y, Wu D, Li S, Liu H, Tian M, Chen Y, Bowie M, Hariharan S, Waitkus M, Keir ST, Sugarman ET, Deek RA, Labrie M, Khasraw M, Lu Y, Mills GB, Herlyn M, Wu K, Liu L, Wei Z, Flaherty KT, Abdullah K, Zhang G, and Ashley DM
- Subjects
- Animals, Cell Line, Tumor, Humans, Mice, Mitochondria metabolism, Temozolomide pharmacology, Temozolomide therapeutic use, Xenograft Model Antitumor Assays, Antineoplastic Agents therapeutic use, Brain Neoplasms drug therapy, Brain Neoplasms genetics, Brain Neoplasms pathology, Glioma drug therapy, Glioma genetics, Glioma metabolism
- Abstract
Purpose: To investigate the antitumor activity of a mitochondrial-localized HSP90 inhibitor, Gamitrinib, in multiple glioma models, and to elucidate the antitumor mechanisms of Gamitrinib in gliomas., Experimental Design: A broad panel of primary and temozolomide (TMZ)-resistant human glioma cell lines were screened by cell viability assays, flow cytometry, and crystal violet assays to investigate the therapeutic efficacy of Gamitrinib. Seahorse assays were used to measure the mitochondrial respiration of glioma cells. Integrated analyses of RNA sequencing (RNAseq) and reverse phase protein array (RPPA) data were performed to reveal the potential antitumor mechanisms of Gamitrinib. Neurospheres, patient-derived organoids (PDO), cell line-derived xenografts (CDX), and patient-derived xenografts (PDX) models were generated to further evaluate the therapeutic efficacy of Gamitrinib., Results: Gamitrinib inhibited cell proliferation and induced cell apoptosis and death in 17 primary glioma cell lines, 6 TMZ-resistant glioma cell lines, 4 neurospheres, and 3 PDOs. Importantly, Gamitrinib significantly delayed the tumor growth and improved survival of mice in both CDX and PDX models in which tumors were either subcutaneously or intracranially implanted. Integrated computational analyses of RNAseq and RPPA data revealed that Gamitrinib exhibited its antitumor activity via (i) suppressing mitochondrial biogenesis, OXPHOS, and cell-cycle progression and (ii) activating the energy-sensing AMP-activated kinase, DNA damage, and stress response., Conclusions: These preclinical findings established the therapeutic role of Gamitrinib in gliomas and revealed the inhibition of mitochondrial biogenesis and tumor bioenergetics as the primary antitumor mechanisms in gliomas., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
314. Epigenetic STING silencing is developmentally conserved in gliomas and can be rescued by methyltransferase inhibition.
- Author
-
Low JT, Chandramohan V, Bowie ML, Brown MC, Waitkus MS, Briley A, Stevenson K, Fuller R, Reitman ZJ, Muscat AM, Hariharan S, Hostettler J, Danehower S, Baker A, Khasraw M, Wong NC, Gregory S, Nair SK, Heimberger A, Gromeier M, Bigner DD, and Ashley DM
- Subjects
- Epigenesis, Genetic, Humans, Glioma drug therapy, Glioma genetics, Methyltransferases
- Abstract
Competing Interests: Declaration of interests The authors declare no competing interests.
- Published
- 2022
- Full Text
- View/download PDF
315. Designing Clinical Trials for Combination Immunotherapy: A Framework for Glioblastoma.
- Author
-
Singh K, Batich KA, Wen PY, Tan AC, Bagley SJ, Lim M, Platten M, Colman H, Ashley DM, Chang SM, Rahman R, Galanis E, Mansouri A, Puduvalli VK, Reardon DA, Sahebjam S, Sampson JH, Simes J, Berry DA, Zadeh G, Cloughesy TF, Mehta MP, Piantadosi S, Weller M, Heimberger AB, and Khasraw M
- Subjects
- Humans, Immune Tolerance, Immunosuppression Therapy, Immunotherapy, Brain Neoplasms pathology, Glioblastoma pathology
- Abstract
Immunotherapy has revolutionized treatment for many hard-to-treat cancers but has yet to produce significant improvement in outcomes for patients with glioblastoma. This reflects the multiple and unique mechanisms of immune evasion and escape in this highly heterogeneous tumor. Glioblastoma engenders profound local and systemic immunosuppression and is remarkably effective at inducing T-cell dysfunction, posing a challenge to any immunotherapy-based approach. To overcome these mechanisms, multiple disparate modes of immune-oriented therapy will be required. However, designing trials that can evaluate these combinatorial approaches requires careful consideration. In this review, we explore the immunotherapy resistance mechanisms that have been encountered to date and how combinatorial approaches may address these. We also describe the unique aspects of trial design in both preclinical and clinical settings and consider endpoints and markers of response best suited for an intervention involving multiple agents., (©2021 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
316. A Modified Nucleoside 6-Thio-2'-Deoxyguanosine Exhibits Antitumor Activity in Gliomas.
- Author
-
Yu S, Wei S, Savani M, Lin X, Du K, Mender I, Siteni S, Vasilopoulos T, Reitman ZJ, Ku Y, Wu D, Liu H, Tian M, Chen Y, Labrie M, Charbonneau CM, Sugarman E, Bowie M, Hariharan S, Waitkus M, Jiang W, McLendon RE, Pan E, Khasraw M, Walsh KM, Lu Y, Herlyn M, Mills G, Herbig U, Wei Z, Keir ST, Flaherty K, Liu L, Wu K, Shay JW, Abdullah K, Zhang G, and Ashley DM
- Subjects
- Animals, Cell Line, Tumor, Deoxyguanosine analogs & derivatives, Humans, Mice, Nucleosides therapeutic use, Proteomics, Thionucleosides, Xenograft Model Antitumor Assays, Brain Neoplasms drug therapy, Brain Neoplasms genetics, Brain Neoplasms pathology, Glioma drug therapy, Glioma genetics, Glioma pathology
- Abstract
Purpose: To investigate the therapeutic role of a novel telomere-directed inhibitor, 6-thio-2'-deoxyguanosine (THIO) in gliomas both in vitro and in vivo ., Experimental Design: A panel of human and mouse glioma cell lines was used to test therapeutic efficacy of THIO using cell viability assays, flow cytometric analyses, and immunofluorescence. Integrated analyses of RNA sequencing and reverse-phase protein array data revealed the potential antitumor mechanisms of THIO. Four patient-derived xenografts (PDX), two patient-derived organoids (PDO), and two xenografts of human glioma cell lines were used to further investigate the therapeutic efficacy of THIO., Results: THIO was effective in the majority of human and mouse glioma cell lines with no obvious toxicity against normal astrocytes. THIO as a monotherapy demonstrated efficacy in three glioma cell lines that had acquired resistance to temozolomide. In addition, THIO showed efficacy in four human glioma cell lines grown as neurospheres by inducing apoptotic cell death. Mechanistically, THIO induced telomeric DNA damage not only in glioma cell lines but also in PDX tumor specimens. Integrated computational analyses of transcriptomic and proteomic data indicated that THIO significantly inhibited cell invasion, stem cell, and proliferation pathways while triggering DNA damage and apoptosis. Importantly, THIO significantly decreased tumor proliferation in two PDO models and reduced the tumor size of a glioblastoma xenograft and a PDX model., Conclusions: The current study established the therapeutic role of THIO in primary and recurrent gliomas and revealed the acute induction of telomeric DNA damage as a primary antitumor mechanism of THIO in gliomas., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
317. Systematic review of combinations of targeted or immunotherapy in advanced solid tumors.
- Author
-
Tan AC, Bagley SJ, Wen PY, Lim M, Platten M, Colman H, Ashley DM, Wick W, Chang SM, Galanis E, Mansouri A, Khagi S, Mehta MP, Heimberger AB, Puduvalli VK, Reardon DA, Sahebjam S, Simes J, Antonia SJ, Berry D, and Khasraw M
- Subjects
- Humans, Combined Modality Therapy methods, Immunotherapy methods, Neoplasms drug therapy
- Abstract
With rapid advances in our understanding of cancer, there is an expanding number of potential novel combination therapies, including novel-novel combinations. Identifying which combinations are appropriate and in which subpopulations are among the most difficult questions in medical research. We conducted a Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA)-guided systematic review of trials of novel-novel combination therapies involving immunotherapies or molecular targeted therapies in advanced solid tumors. A MEDLINE search was conducted using a modified Cochrane Highly Sensitive Search Strategy for published clinical trials between July 1, 2017, and June 30, 2020, in the top-ranked medical and oncology journals. Trials were evaluated according to a criterion adapted from previously published Food and Drug Administration guidance and other key considerations in designing trials of combinations. This included the presence of a strong biological rationale, the use of a new established or emerging predictive biomarker prospectively incorporated into the clinical trial design, appropriate comparator arms of monotherapy or supportive external data sources and a primary endpoint demonstrating a clinically meaningful benefit. Of 32 identified trials, there were 11 (34%) trials of the novel-novel combination of anti-programmed death 1 (PD-1)/programmed death ligand 1 (PD-L1) and anti-cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) therapy, and 10 (31%) trials of anti-PD-1/PD-L1 and anti-vascular endothelial growth factor (VEGF) combination therapy. 20 (62.5%) trials were phase II trials, while 12 (37.5%) were phase III trials. Most (72%) trials lacked significant preclinical evidence supporting the development of the combination in the given indication. A majority of trials (69%) were conducted in biomarker unselected populations or used pre-existing biomarkers within the given indication for patient selection. Most studies (66%) were considered to have appropriate comparator arms or had supportive external data sources such as prior studies of monotherapy. All studies were evaluated as selecting a clinically meaningful primary endpoint. In conclusion, designing trials to evaluate novel-novel combination therapies presents numerous challenges to demonstrate efficacy in a comprehensive manner. A greater understanding of biological rationale for combinations and incorporating predictive biomarkers may improve effective evaluation of combination therapies. Innovative statistical methods and increasing use of external data to support combination approaches are potential strategies that may improve the efficiency of trial design. Designing trials to evaluate novel-novel combination therapies presents numerous challenges to demonstrate efficacy in a comprehensive manner. A greater understanding of biological rationale for combinations and incorporating predictive biomarkers may improve effective evaluation of combination therapies. Innovative statistical methods and increasing use of external data to support combination approaches are potential strategies that may improve the efficiency of trial design., Competing Interests: Competing interests: ACT reports consultant or advisory roles for Amgen. SJB reports research grants from Incyte, Eli Lilly, Novocure, GSK; consultant or advisory roles for Bayer and Novocure; and US patent for 'Combination therapies of EGFRvIII chimeric antigen receptors and PD-1 inhibitors'. PYW reports consultant or advisory roles for Agios, Astra Zeneca, Bayer, Boston Pharmaceuticals, CNS Pharmaceuticals, Elevate Bio Immunomic Therapeutics, Imvax, Karyopharm, Merck, Novartis, Nuvation Bio, Vascular Biogenics, VBI Vaccines, Voyager and QED; research funding from Agios, Astra Zeneca/Medimmune, Beigene, Celgene, Eli Lily, Genentech/Roche, Kazia, MediciNova, Merck, Novartis, Nuvation Bio, Oncoceutics, Vascular Biogenics and VBI Vaccines; is an editor for UpToDate and Elsevier. ML reports consultant or advisory roles for Tocagen, SQZ Technologies, VBI, InCephalo Therapeutics and Pyramid Bio; non-research consulting roles for Stryker; research support from Arbor, Bristol-Myers Squibb, Accuray, DNAtrix, Tocagen, Biohaven and Kyrin-Kyowa; and has patents for focused radiation+checkpoint inhibitors and local chemotherapy+checkpoint inhibitors. MP reports consultant or advisory roles for non-financial support from Roche, personal fees and non-financial support from Bayer, personal fees from Novartis, personal fees from Apogenix, non-financial support from Pfizer, personal fees from Affiris outside the submitted work. In addition, MP has a patent EP2753315B1 licensed to Bayer, a patent EP2800580B1 issued, a patent US20180155403A1 pending, a patent US20180246118A1 pending, a patent US20170254803A1 pending, a patent WO2018146010A1 licensed to Bayer, a patent WO2019101643A1 licensed to Bayer, a patent WO2019101647A1 licensed to Bayer, a patent WO2019101641A1 licensed to Bayer, and a patent WO2019101642A1 licensed to Bayer. HC reports consultant or advisory roles for Best Doctors/Teladoc, Karyopharm Therapeutics, Private Health, Orbus, Bayer, Forma Therapeutics and Adastra Pharmaceuticals; research funding (Inst) from Newlink Genetics, Plexxikon, Kadmon, Orbus, Merck, DNATrix, Abbvie, Beigene, Forma Therapeutics, GCAR, Array BioPharma, Karyopharm, Nuvation Bio, Bayer and Bristol Meyer Squibb. DMA reports stock and other ownership interests in Diverse Biotech; consulting or advisory roles for Istari Oncology and Jackson Laboratory for Genomic Medicine; patents, royalties, other intellectual property for 'Methods for predicting tumor response to immunotherapy, U.S. Provisional application no. 62/787' and 'Methods for predicting tumor response to immunotherapy, U.S. Provisional application no. 62/620,577'; and expert testimony for Tanoury, Nauts, McKinney & Gabarino, PLLC. WW reports consultant or advisory roles (Inst) for Agios, Bayer, MSD, Novartis and Roche; research funding (Inst) from Apogenix, Boehringer Ingelheim, Pfizer and Roche. SMC reports research funding (Inst) from Agios. EG reports consultant or advisory roles (Inst) for MedImmune, Inc., Agios Pharmaceuticals and Karyopharm; consultant or advisory roles for Gradalis, Inc. and Kiyatec, Inc.; research funding (Inst) from MedImmune, Inc., Tracon, Genentech and Bristol-Myers Squibb. MPM reports consultant or advisory roles for Zap, Mevion, Karyopharm, Tocagen and Astra-Zeneca; and Board of Directors options from Oncoceutics. ABH reports consultant or advisory roles for Caris Life Sciences and WCG Oncology; royalties on licensed intellectual property from Celldex Therapeutics and DNAtrix; research funding from Celularity, Carthera, Codiak, and Moleculin. DAR reports consultant or advisory roles for Abbvie, Advantagene, Agenus, Amgen, Bayer, Bristol-Myers Squibb, Celldex, DelMar, EMD Serono, Genentech/Roche, Imvax, Inovio, Medicenna Biopharma, Inc., Merck, Merck KGaA, Monteris, Novocure, Oncorus, Oxigene, Regeneron, Stemline, Sumitono Dainippon Pharma and Taiho Oncology, Inc.; honoraria from Abbvie, Advantagene, Agenus, Bristol-Myers Squibb, Celldex, EMD Serono, Genentech/Roche, Imvax, Inovio, Medicenna Biopharma, Inc., Merck, Merck KGaA, Monteris, Novocure, Oncorus, Oxigene, Regeneron, Stemline, Sumitono Dainippon Pharma and Taiho Oncology, Inc.; research funding (Inst) from Acerta Phamaceuticals, Agenus, Celldex, EMD Serono, Incyte, Inovio, Omniox and Tragara. SS reports consultant or advisory roles for Merck and Boehringer Ingelheim; research funding from Merck, Bristol-Myers Squibb, and Brooklyn ImmunoTherapeutics. RJS reports research funding (Inst) from Bayer, Astra Zeneca, Pfizer, Astella, Roche, Merck, Abbvie and Amgen. SJA reports consultant or advisory roles for Achilles, Amgen, AstraZeneca, Bristol-Myers Squibb, Caris Life Sciences, CBMG, Celsius Therapeutics, G1 Therapeutics, GlaxoSmithKline, Memgen, Merck, Nektar, RAPT Therapeutics, Venn, Glympse and Samyang; data review committee for EMD Serano. DB is co-owner of Berry Consultants, LLC, a company that designs adaptive Bayesian clinical trials for pharmaceutical and medical device companies, NIH cooperative groups, patient advocacy groups, and international consortia. MK reports consultant or advisory roles for Janssen, AbbVie, Ipsen, Pfizer Roche, and Jackson Laboratory for Genomic Medicine; research funding from AbbVie, Bristol-Myers Squibb, and Specialized Therapeutics. AM, SK and VKP report no conflicts of interest., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2021
- Full Text
- View/download PDF
318. Viral infection of cells within the tumor microenvironment mediates antitumor immunotherapy via selective TBK1-IRF3 signaling.
- Author
-
Brown MC, Mosaheb MM, Mohme M, McKay ZP, Holl EK, Kastan JP, Yang Y, Beasley GM, Hwang ES, Ashley DM, Bigner DD, Nair SK, and Gromeier M
- Subjects
- Animals, Breast Neoplasms immunology, Breast Neoplasms pathology, CD8-Positive T-Lymphocytes immunology, Female, Humans, Immunity, Innate immunology, Interferon Type I immunology, Interferon-Induced Helicase, IFIH1 metabolism, Lymphocytes, Tumor-Infiltrating immunology, Melanoma immunology, Melanoma pathology, Mice, Mice, Inbred C57BL, Mice, Knockout, NF-kappa B metabolism, Signal Transduction immunology, Th1 Cells immunology, Breast Neoplasms therapy, Interferon Regulatory Factor-3 immunology, Melanoma therapy, Oncolytic Virotherapy, Protein Serine-Threonine Kinases immunology, Tumor Microenvironment immunology
- Abstract
Activating intra-tumor innate immunity might enhance tumor immune surveillance. Virotherapy is proposed to achieve tumor cell killing, while indirectly activating innate immunity. Here, we report that recombinant poliovirus therapy primarily mediates antitumor immunotherapy via direct infection of non-malignant tumor microenvironment (TME) cells, independent of malignant cell lysis. Relative to other innate immune agonists, virotherapy provokes selective, TBK1-IRF3 driven innate inflammation that is associated with sustained type-I/III interferon (IFN) release. Despite priming equivalent antitumor T cell quantities, MDA5-orchestrated TBK1-IRF3 signaling, but not NFκB-polarized TLR activation, culminates in polyfunctional and Th1-differentiated antitumor T cell phenotypes. Recombinant type-I IFN increases tumor-localized T cell function, but does not mediate durable antitumor immunotherapy without concomitant pattern recognition receptor (PRR) signaling. Thus, virus-induced MDA5-TBK1-IRF3 signaling in the TME provides PRR-contextualized IFN responses that elicit functional antitumor T cell immunity. TBK1-IRF3 innate signal transduction stimulates eventual function and differentiation of tumor-infiltrating T cells.
- Published
- 2021
- Full Text
- View/download PDF
319. Very low mutation burden is a feature of inflamed recurrent glioblastomas responsive to cancer immunotherapy.
- Author
-
Gromeier M, Brown MC, Zhang G, Lin X, Chen Y, Wei Z, Beaubier N, Yan H, He Y, Desjardins A, Herndon JE 2nd, Varn FS, Verhaak RG, Zhao J, Bolognesi DP, Friedman AH, Friedman HS, McSherry F, Muscat AM, Lipp ES, Nair SK, Khasraw M, Peters KB, Randazzo D, Sampson JH, McLendon RE, Bigner DD, and Ashley DM
- Subjects
- Biomarkers, Tumor genetics, Brain Neoplasms genetics, Brain Neoplasms pathology, Cohort Studies, Gene Expression Profiling methods, Gene Expression Regulation, Neoplastic, Genomics methods, Glioblastoma genetics, Glioblastoma pathology, Humans, Inflammation genetics, Neoplasm Recurrence, Local, Outcome Assessment, Health Care methods, Outcome Assessment, Health Care statistics & numerical data, Proportional Hazards Models, Survival Analysis, Brain Neoplasms therapy, Glioblastoma therapy, Immunotherapy methods, Mutation, Oncolytic Virotherapy methods
- Abstract
Several immunotherapy clinical trials in recurrent glioblastoma have reported long-term survival benefits in 10-20% of patients. Here we perform genomic analysis of tumor tissue from recurrent WHO grade IV glioblastoma patients acquired prior to immunotherapy intervention. We report that very low tumor mutation burden is associated with longer survival after recombinant polio virotherapy or after immune checkpoint blockade in recurrent glioblastoma patients. A relationship between tumor mutation burden and survival is not observed in cohorts of immunotherapy naïve newly diagnosed or recurrent glioblastoma patients. Transcriptomic analyses reveal an inverse relationship between tumor mutation burden and enrichment of inflammatory gene signatures in cohorts of recurrent, but not newly diagnosed glioblastoma tumors, implying that a relationship between tumor mutation burden and tumor-intrinsic inflammation evolves upon recurrence.
- Published
- 2021
- Full Text
- View/download PDF
320. Genetically stable poliovirus vectors activate dendritic cells and prime antitumor CD8 T cell immunity.
- Author
-
Mosaheb MM, Dobrikova EY, Brown MC, Yang Y, Cable J, Okada H, Nair SK, Bigner DD, Ashley DM, and Gromeier M
- Subjects
- Animals, Cancer Vaccines, Genetic Vectors immunology, Glioma immunology, HEK293 Cells, HeLa Cells, Humans, Immunity, Innate, Immunotherapy methods, Interferon Type I immunology, Melanoma immunology, Mice, Mice, Inbred C57BL, Poliovirus genetics, CD8-Positive T-Lymphocytes physiology, Dendritic Cells immunology, Poliovirus immunology
- Abstract
Viruses naturally engage innate immunity, induce antigen presentation, and mediate CD8 T cell priming against foreign antigens. Polioviruses can provide a context optimal for generating antigen-specific CD8 T cells, as they have natural tropism for dendritic cells, preeminent inducers of CD8 T cell immunity; elicit Th1-promoting inflammation; and lack interference with innate or adaptive immunity. However, notorious genetic instability and underlying neuropathogenicity has hampered poliovirus-based vector applications. Here we devised a strategy based on the polio:rhinovirus chimera PVSRIPO, devoid of viral neuropathogenicity after intracerebral inoculation in human subjects, for stable expression of exogenous antigens. PVSRIPO vectors infect, activate, and induce epitope presentation in DCs in vitro; they recruit and activate DCs with Th1-dominant cytokine profiles at the injection site in vivo. They efficiently prime tumor antigen-specific CD8 T cells in vivo, induce CD8 T cell migration to the tumor site, delay tumor growth and enhance survival in murine tumor models.
- Published
- 2020
- Full Text
- View/download PDF
321. Brain immunology and immunotherapy in brain tumours.
- Author
-
Sampson JH, Gunn MD, Fecci PE, and Ashley DM
- Subjects
- Animals, Biomarkers, Tumor, Blood-Brain Barrier metabolism, Brain Neoplasms metabolism, Brain Neoplasms pathology, Clinical Trials as Topic, Combined Modality Therapy, Disease Management, Disease Susceptibility, Humans, Immunomodulation, Standard of Care, Treatment Outcome, Tumor Microenvironment, Brain immunology, Brain metabolism, Brain Neoplasms etiology, Brain Neoplasms therapy, Immunity, Immunotherapy
- Abstract
Gliomas, the most common malignant primary brain tumours, remain universally lethal. Yet, seminal discoveries in the past 5 years have clarified the anatomy, genetics and function of the immune system within the central nervous system (CNS) and altered the paradigm for successful immunotherapy. The impact of standard therapies on the response to immunotherapy is now better understood, as well. This new knowledge has implications for a broad range of tumours that develop within the CNS. Nevertheless, the requirements for successful therapy remain effective delivery and target specificity, while the dramatic heterogeneity of malignant gliomas at the genetic and immunological levels remains a profound challenge.
- Published
- 2020
- Full Text
- View/download PDF
322. Longitudinal molecular trajectories of diffuse glioma in adults.
- Author
-
Barthel FP, Johnson KC, Varn FS, Moskalik AD, Tanner G, Kocakavuk E, Anderson KJ, Abiola O, Aldape K, Alfaro KD, Alpar D, Amin SB, Ashley DM, Bandopadhayay P, Barnholtz-Sloan JS, Beroukhim R, Bock C, Brastianos PK, Brat DJ, Brodbelt AR, Bruns AF, Bulsara KR, Chakrabarty A, Chakravarti A, Chuang JH, Claus EB, Cochran EJ, Connelly J, Costello JF, Finocchiaro G, Fletcher MN, French PJ, Gan HK, Gilbert MR, Gould PV, Grimmer MR, Iavarone A, Ismail A, Jenkinson MD, Khasraw M, Kim H, Kouwenhoven MCM, LaViolette PS, Li M, Lichter P, Ligon KL, Lowman AK, Malta TM, Mazor T, McDonald KL, Molinaro AM, Nam DH, Nayyar N, Ng HK, Ngan CY, Niclou SP, Niers JM, Noushmehr H, Noorbakhsh J, Ormond DR, Park CK, Poisson LM, Rabadan R, Radlwimmer B, Rao G, Reifenberger G, Sa JK, Schuster M, Shaw BL, Short SC, Smitt PAS, Sloan AE, Smits M, Suzuki H, Tabatabai G, Van Meir EG, Watts C, Weller M, Wesseling P, Westerman BA, Widhalm G, Woehrer A, Yung WKA, Zadeh G, Huse JT, De Groot JF, Stead LF, and Verhaak RGW
- Subjects
- Adult, Chromosomes, Human, Pair 1, Chromosomes, Human, Pair 19, Disease Progression, Glioma pathology, Humans, Isocitrate Dehydrogenase genetics, Mutation, Polymorphism, Single Nucleotide, Recurrence, Glioma genetics
- Abstract
The evolutionary processes that drive universal therapeutic resistance in adult patients with diffuse glioma remain unclear
1,2 . Here we analysed temporally separated DNA-sequencing data and matched clinical annotation from 222 adult patients with glioma. By analysing mutations and copy numbers across the three major subtypes of diffuse glioma, we found that driver genes detected at the initial stage of disease were retained at recurrence, whereas there was little evidence of recurrence-specific gene alterations. Treatment with alkylating agents resulted in a hypermutator phenotype at different rates across the glioma subtypes, and hypermutation was not associated with differences in overall survival. Acquired aneuploidy was frequently detected in recurrent gliomas and was characterized by IDH mutation but without co-deletion of chromosome arms 1p/19q, and further converged with acquired alterations in the cell cycle and poor outcomes. The clonal architecture of each tumour remained similar over time, but the presence of subclonal selection was associated with decreased survival. Finally, there were no differences in the levels of immunoediting between initial and recurrent gliomas. Collectively, our results suggest that the strongest selective pressures occur during early glioma development and that current therapies shape this evolution in a largely stochastic manner.- Published
- 2019
- Full Text
- View/download PDF
323. Complementary and integrative health interventions and their association with health-related quality of life in the primary brain tumor population.
- Author
-
Randazzo DM, McSherry F, Herndon JE, Affronti ML, Lipp ES, Flahiff C, Miller E, Woodring S, Boulton S, Desjardins A, Ashley DM, Friedman HS, and Peters KB
- Subjects
- Humans, Retrospective Studies, Brain Neoplasms epidemiology, Brain Neoplasms therapy, Complementary Therapies statistics & numerical data, Quality of Life
- Abstract
Background and Purpose: Little is known about complementary and integrative health intervention usage in the primary brain tumor population. We aimed to identify the percentage of patients using these practices and explore the impact on quality of life., Materials and Methods: Clinical records from patients seen in clinic between December 16, 2013 and February 28, 2014 were reviewed retrospectively. The questionnaires used were a modified version of the International Complementary and Alternative Medicine Questionnaire, the Functional Assessment of Cancer Therapy- Brain Cancer and the Functional Assessment of Chronic Illness Therapy- Fatigue., Results: 76% of patients utilized a complementary and integrative health modality. The most frequently reported modalities used were vitamins, massage, and spiritual healing, prayer, diet and meditation., Conclusion: These results confirm the usage of complementary and integrative health practices within the primary brain tumor population; however, there was no evidence of association between use and quality of life., (Copyright © 2019. Published by Elsevier Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
324. Human Bcl-2 cannot directly inhibit the Caenorhabditis elegans Apaf-1 homologue CED-4, but can interact with EGL-1.
- Author
-
Jabbour AM, Puryer MA, Yu JY, Lithgow T, Riffkin CD, Ashley DM, Vaux DL, Ekert PG, and Hawkins CJ
- Subjects
- Animals, Animals, Genetically Modified, Apoptosis physiology, Apoptosis Regulatory Proteins metabolism, Caenorhabditis elegans, Humans, Protein Binding, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-bcl-2 pharmacology, Saccharomyces cerevisiae metabolism, Two-Hybrid System Techniques, bcl-2-Associated X Protein antagonists & inhibitors, Caenorhabditis elegans Proteins metabolism, Calcium-Binding Proteins metabolism, Proto-Oncogene Proteins c-bcl-2 physiology, Repressor Proteins metabolism
- Abstract
Although the anti-apoptotic activity of Bcl-2 has been extensively studied, its mode of action is still incompletely understood. In the nematode Caenorhabditis elegans, 131 of 1090 somatic cells undergo programmed cell death during development. Transgenic expression of human Bcl-2 reduced cell death during nematode development, and partially complemented mutation of ced-9, indicating that Bcl-2 can functionally interact with the nematode cell death machinery. Identification of the nematode target(s) of Bcl-2 inhibition would help clarify the mechanism by which Bcl-2 suppresses apoptosis in mammalian cells. Exploiting yeast-based systems and biochemical assays, we analysed the ability of Bcl-2 to interact with and regulate the activity of nematode apoptosis proteins. Unlike CED-9, Bcl-2 could not directly associate with the caspase-activating adaptor protein CED-4, nor could it inhibit CED-4-dependent yeast death. By contrast, Bcl-2 could bind the C. elegans pro-apoptotic BH3-only Bcl-2 family member EGL-1. These data prompt us to hypothesise that Bcl-2 might suppress nematode cell death by preventing EGL-1 from antagonising CED-9, rather than by inhibiting CED-4.
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.