28 results on '"Cahill DP"'
Search Results
2. Mutant IDH Modulates Suppressive Myeloid Populations in Malignant Glioma.
- Author
-
Grewal EP, Richardson LGK, Sun J, Ramapriyan R, Martinez-Lage M, Miller JJ, Carter BS, Cahill DP, Curry WT, and Choi BD
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Myeloid Cells pathology, Myeloid Cells metabolism, Gene Expression Regulation, Neoplastic, Disease Models, Animal, Transcriptome, Isocitrate Dehydrogenase genetics, Mutation, Glioma genetics, Glioma pathology, Myeloid-Derived Suppressor Cells metabolism, Brain Neoplasms genetics, Brain Neoplasms pathology
- Abstract
Purpose: Mutations in the isocitrate dehydrogenase (IDH) genes IDH1 and IDH2 have critical diagnostic and prognostic significance in diffuse gliomas. Neomorphic mutant IDH activity has been previously implicated in T-cell suppression; however, the effects of IDH mutations on intratumoral myeloid populations remain underexplored. In this study, we investigate the influence of IDH status on the myeloid compartment using human glioma specimens and preclinical models., Experimental Design: We performed RNA sequencing and quantitative immunofluorescence on newly diagnosed, treatment-naive IDH-mutant grade 4 astrocytoma and IDH-wild-type (IDH-WT) glioblastoma (GBM) specimens. We also generated a syngeneic murine model, comparing transcriptomic and cell-level changes in paired isogenic glioma lines that differ only in IDH mutational status., Results: Among patient samples, IDH-mutant tumors displayed an underrepresentation of suppressive myeloid transcriptional signatures, which was confirmed at the cellular level with decreased numbers of intratumoral M2-like macrophages and myeloid-derived suppressor cells. Introduction of the mutant IDH enzyme into murine glioma was sufficient to recapitulate the transcriptomic and cellular shifts observed in patient samples., Conclusions: We provide transcriptomic and cellular evidence that mutant IDH is associated with a quantitative reduction of suppressive myeloid cells in gliomas and that introduction of the mutant enzyme is sufficient to result in corresponding cellular changes using an in vivo preclinical model. These data advance our understanding of high-grade gliomas by identifying key myeloid cell populations that are reprogrammed by mutant IDH and may be targetable through therapeutic approaches., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
3. CDKN2A/B Homozygous Deletion Sensitizes IDH-Mutant Glioma to CDK4/6 Inhibition.
- Author
-
Nasser AM, Melamed L, Wetzel EA, Chang JC, Nagashima H, Kitagawa Y, Muzyka L, Wakimoto H, Cahill DP, and Miller JJ
- Subjects
- Humans, Animals, Mice, Cell Line, Tumor, Gene Deletion, Brain Neoplasms genetics, Brain Neoplasms drug therapy, Brain Neoplasms pathology, Homozygote, Cyclin-Dependent Kinase 4 antagonists & inhibitors, Cyclin-Dependent Kinase 4 genetics, Cyclin-Dependent Kinase 6 antagonists & inhibitors, Cyclin-Dependent Kinase 6 genetics, Glioma genetics, Glioma drug therapy, Glioma pathology, Cyclin-Dependent Kinase Inhibitor p16 genetics, Isocitrate Dehydrogenase genetics, Isocitrate Dehydrogenase antagonists & inhibitors, Xenograft Model Antitumor Assays, Benzimidazoles pharmacology, Cyclin-Dependent Kinase Inhibitor p15 genetics, Aminopyridines pharmacology, Mutation, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Piperazines pharmacology, Piperazines therapeutic use, Cell Proliferation drug effects, Pyridines pharmacology, Pyridines therapeutic use
- Abstract
Purpose: Treatment paradigms for isocitrate dehydrogenase (IDH)-mutant gliomas are rapidly evolving. Although typically indolent and responsive to initial treatment, these tumors invariably recur at a higher grade and require salvage treatment. Homozygous deletion of the tumor suppressor gene CDKN2A/B frequently emerges at recurrence in these tumors, driving poor patient outcomes. We investigated the effect of CDK-Rb pathway blockade on IDH-mutant glioma growth in vitro and in vivo using CDK4/6 inhibitors (CDKi)., Experimental Design: Cell viability, proliferation assays, and flow cytometry were used to examine the pharmacologic effect of two distinct CDKi, palbociclib and abemaciclib, in multiple patient-derived IDH-mutant glioma lines. Isogenic models were used to directly investigate the influence of CDKN2A/B status on CDKi sensitivity. Orthotopic xenograft tumor models were used to examine the efficacy and tolerability of CDKi in vivo., Results: CDKi treatment leads to decreased cell viability and proliferative capacity in patient-derived IDH-mutant glioma lines, coupled with enrichment of cells in the G1 phase. CDKN2A inactivation sensitizes IDH-mutant glioma to CDKi in both endogenous and isogenic models with engineered CDKN2A deletion. CDK4/6 inhibitor administration improves survival in orthotopically implanted IDH-mutant glioma models., Conclusions: IDH-mutant gliomas with deletion of CDKN2A/B are sensitized to CDK4/6 inhibitors. These results support the investigation of the use of these agents in a clinical setting., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
4. Glioblastoma-Infiltrating CD8+ T Cells Are Predominantly a Clonally Expanded GZMK+ Effector Population.
- Author
-
Wang AZ, Mashimo BL, Schaettler MO, Sherpa ND, Leavitt LA, Livingstone AJ, Khan SM, Li M, Anzaldua-Campos MI, Bradley JD, Leuthardt EC, Kim AH, Dowling JL, Chicoine MR, Jones PS, Choi BD, Cahill DP, Carter BS, Petti AA, Johanns TM, and Dunn GP
- Subjects
- Humans, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism, Tumor Microenvironment immunology, Brain Neoplasms immunology, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Glioblastoma immunology, Glioblastoma therapy
- Abstract
Recent clinical trials have highlighted the limited efficacy of T cell-based immunotherapy in patients with glioblastoma (GBM). To better understand the characteristics of tumor-infiltrating lymphocytes (TIL) in GBM, we performed cellular indexing of transcriptomes and epitopes by sequencing and single-cell RNA sequencing with paired V(D)J sequencing, respectively, on TILs from two cohorts of patients totaling 15 patients with high-grade glioma, including GBM or astrocytoma, IDH-mutant, grade 4 (G4A). Analysis of the CD8+ TIL landscape reveals an enrichment of clonally expanded GZMK+ effector T cells in the tumor compared with matched blood, which was validated at the protein level. Furthermore, integration with other cancer types highlights the lack of a canonically exhausted CD8+ T-cell population in GBM TIL. These data suggest that GZMK+ effector T cells represent an important T-cell subset within the GBM microenvironment and may harbor potential therapeutic implications., Significance: To understand the limited efficacy of immune-checkpoint blockade in GBM, we applied a multiomics approach to understand the TIL landscape. By highlighting the enrichment of GZMK+ effector T cells and the lack of exhausted T cells, we provide a new potential mechanism of resistance to immunotherapy in GBM. This article is featured in Selected Articles from This Issue, p. 897., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
5. Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors.
- Author
-
Hayashi T, Tateishi K, Matsuyama S, Iwashita H, Miyake Y, Oshima A, Honma H, Sasame J, Takabayashi K, Sugino K, Hirata E, Udaka N, Matsushita Y, Kato I, Hayashi H, Nakamura T, Ikegaya N, Takayama Y, Sonoda M, Oka C, Sato M, Isoda M, Kato M, Uchiyama K, Tanaka T, Muramatsu T, Miyake S, Suzuki R, Takadera M, Tatezuki J, Ayabe J, Suenaga J, Matsunaga S, Miyahara K, Manaka H, Murata H, Yokoyama T, Tanaka Y, Shuto T, Ichimura K, Kato S, Yamanaka S, Cahill DP, Fujii S, Shankar GM, and Yamamoto T
- Subjects
- Adult, Humans, Retrospective Studies, Reproducibility of Results, Central Nervous System Neoplasms diagnosis, Central Nervous System Neoplasms genetics, Brain Neoplasms diagnosis, Brain Neoplasms genetics, Brain Neoplasms pathology, Glioma diagnosis, Glioma genetics, Glioma surgery
- Abstract
Purpose: The 2021 World Health Organization (WHO) classification of central nervous system (CNS) tumors uses an integrated approach involving histopathology and molecular profiling. Because majority of adult malignant brain tumors are gliomas and primary CNS lymphomas (PCNSL), rapid differentiation of these diseases is required for therapeutic decisions. In addition, diffuse gliomas require molecular information on single-nucleotide variants (SNV), such as IDH1/2. Here, we report an intraoperative integrated diagnostic (i-ID) system to classify CNS malignant tumors, which updates legacy frozen-section (FS) diagnosis through incorporation of a qPCR-based genotyping assay., Experimental Design: FS evaluation, including GFAP and CD20 rapid IHC, was performed on adult malignant CNS tumors. PCNSL was diagnosed through positive CD20 and negative GFAP immunostaining. For suspected glioma, genotyping for IDH1/2, TERT SNV, and CDKN2A copy-number alteration was routinely performed, whereas H3F3A and BRAF SNV were assessed for selected cases. i-ID was determined on the basis of the 2021 WHO classification and compared with the permanent integrated diagnosis (p-ID) to assess its reliability., Results: After retrospectively analyzing 153 cases, 101 cases were prospectively examined using the i-ID system. Assessment of IDH1/2, TERT, H3F3AK27M, BRAFV600E, and CDKN2A alterations with i-ID and permanent genomic analysis was concordant in 100%, 100%, 100%, 100%, and 96.4%, respectively. Combination with FS and intraoperative genotyping assay improved diagnostic accuracy in gliomas. Overall, i-ID matched with p-ID in 80/82 (97.6%) patients with glioma and 18/19 (94.7%) with PCNSL., Conclusions: The i-ID system provides reliable integrated diagnosis of adult malignant CNS tumors., (©2023 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
6. Zinc Finger MYND-Type Containing 8 (ZMYND8) Is Epigenetically Regulated in Mutant Isocitrate Dehydrogenase 1 (IDH1) Glioma to Promote Radioresistance.
- Author
-
Carney SV, Banerjee K, Mujeeb A, Zhu B, Haase S, Varela ML, Kadiyala P, Tronrud CE, Zhu Z, Mukherji D, Gorla P, Sun Y, Tagett R, Núñez FJ, Luo M, Luo W, Ljungman M, Liu Y, Xia Z, Schwendeman A, Qin T, Sartor MA, Costello JF, Cahill DP, Lowenstein PR, and Castro MG
- Subjects
- Humans, MYND Domains, Epigenesis, Genetic, Nuclear Proteins genetics, Nuclear Proteins metabolism, Poly(ADP-ribose) Polymerase Inhibitors, Transcription Factors genetics, Transcription Factors metabolism, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Isocitrate Dehydrogenase metabolism, Glioma genetics, Glioma radiotherapy
- Abstract
Purpose: Mutant isocitrate dehydrogenase 1 (mIDH1) alters the epigenetic regulation of chromatin, leading to a hypermethylation phenotype in adult glioma. This work focuses on identifying gene targets epigenetically dysregulated by mIDH1 to confer therapeutic resistance to ionizing radiation (IR)., Experimental Design: We evaluated changes in the transcriptome and epigenome in a radioresistant mIDH1 patient-derived glioma cell culture (GCC) following treatment with an mIDH1-specific inhibitor, AGI-5198. We identified Zinc Finger MYND-Type Containing 8 (ZMYND8) as a potential target of mIDH1 reprogramming. We suppressed ZMYND8 expression by shRNA knockdown and genetic knockout (KO) in mIDH1 glioma cells and then assessed cellular viability to IR. We assessed the sensitivity of mIDH1 GCCS to pharmacologic inhibition of ZMYND8-interacting partners: HDAC, BRD4, and PARP., Results: Inhibition of mIDH1 leads to an upregulation of gene networks involved in replication stress. We found that the expression of ZMYND8, a regulator of DNA damage response, was decreased in three patient-derived mIDH1 GCCs after treatment with AGI-5198. Knockdown of ZMYND8 expression sensitized mIDH1 GCCs to radiotherapy marked by decreased cellular viability. Following IR, mIDH1 glioma cells with ZMYND8 KO exhibit significant phosphorylation of ATM and sustained γH2AX activation. ZMYND8 KO mIDH1 GCCs were further responsive to IR when treated with either BRD4 or HDAC inhibitors. PARP inhibition further enhanced the efficacy of radiotherapy in ZMYND8 KO mIDH1 glioma cells., Conclusions: These findings indicate the impact of ZMYND8 in the maintenance of genomic integrity and repair of IR-induced DNA damage in mIDH1 glioma. See related commentary by Sachdev et al., p. 1648., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
7. Microenvironmental Landscape of Human Melanoma Brain Metastases in Response to Immune Checkpoint Inhibition.
- Author
-
Alvarez-Breckenridge C, Markson SC, Stocking JH, Nayyar N, Lastrapes M, Strickland MR, Kim AE, de Sauvage M, Dahal A, Larson JM, Mora JL, Navia AW, Klein RH, Kuter BM, Gill CM, Bertalan M, Shaw B, Kaplan A, Subramanian M, Jain A, Kumar S, Danish H, White M, Shahid O, Pauken KE, Miller BC, Frederick DT, Hebert C, Shaw M, Martinez-Lage M, Frosch M, Wang N, Gerstner E, Nahed BV, Curry WT, Carter B, Cahill DP, Boland GM, Izar B, Davies MA, Sharpe AH, Suvà ML, Sullivan RJ, Brastianos PK, and Carter SL
- Subjects
- Humans, Immune Checkpoint Inhibitors, Tumor Microenvironment, Brain Neoplasms, Melanoma
- Abstract
Melanoma-derived brain metastases (MBM) represent an unmet clinical need because central nervous system progression is frequently an end stage of the disease. Immune checkpoint inhibitors (ICI) provide a clinical opportunity against MBM; however, the MBM tumor microenvironment (TME) has not been fully elucidated in the context of ICI. To dissect unique elements of the MBM TME and correlates of MBM response to ICI, we collected 32 fresh MBM and performed single-cell RNA sequencing of the MBM TME and T-cell receptor clonotyping on T cells from MBM and matched blood and extracranial lesions. We observed myeloid phenotypic heterogeneity in the MBM TME, most notably multiple distinct neutrophil states, including an IL8-expressing population that correlated with malignant cell epithelial-to-mesenchymal transition. In addition, we observed significant relationships between intracranial T-cell phenotypes and the distribution of T-cell clonotypes intracranially and peripherally. We found that the phenotype, clonotype, and overall number of MBM-infiltrating T cells were associated with response to ICI, suggesting that ICI-responsive MBMs interact with peripheral blood in a manner similar to extracranial lesions. These data identify unique features of the MBM TME that may represent potential targets to improve clinical outcomes for patients with MBM., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
8. HSP90 Inhibition Overcomes Resistance to Molecular Targeted Therapy in BRAFV600E-mutant High-grade Glioma.
- Author
-
Sasame J, Ikegaya N, Kawazu M, Natsumeda M, Hayashi T, Isoda M, Satomi K, Tomiyama A, Oshima A, Honma H, Miyake Y, Takabayashi K, Nakamura T, Ueno T, Matsushita Y, Iwashita H, Kanemaru Y, Murata H, Ryo A, Terashima K, Yamanaka S, Fujii Y, Mano H, Komori T, Ichimura K, Cahill DP, Wakimoto H, Yamamoto T, and Tateishi K
- Subjects
- Cell Line, Tumor, Drug Resistance, Neoplasm genetics, Humans, Mitogen-Activated Protein Kinase Kinases antagonists & inhibitors, Mitogen-Activated Protein Kinase Kinases metabolism, Molecular Targeted Therapy, Mutation, Neoplasm Recurrence, Local drug therapy, Protein Kinase Inhibitors, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Proto-Oncogene Proteins B-raf genetics, Proto-Oncogene Proteins B-raf metabolism, Proto-Oncogene Proteins c-akt genetics, Proto-Oncogene Proteins c-akt metabolism, Antineoplastic Combined Chemotherapy Protocols pharmacology, Brain Neoplasms drug therapy, Brain Neoplasms genetics, Brain Neoplasms metabolism, Glioma drug therapy, Glioma genetics, Glioma metabolism, HSP90 Heat-Shock Proteins antagonists & inhibitors, HSP90 Heat-Shock Proteins genetics, Melanoma drug therapy, Melanoma genetics, Melanoma metabolism
- Abstract
Purpose: Molecular targeted therapy using BRAF and/or MEK inhibitors has been applied to BRAFV600E-mutant high-grade gliomas (HGG); however, the therapeutic effect is limited by the emergence of drug resistance., Experimental Design: We established multiple paired BRAFV600E-mutant HGG patient-derived xenograft models based on tissues collected prior to and at relapse after molecular targeted therapy. Using these models, we dissected treatment-resistant mechanisms for molecular targeted therapy and explored therapeutic targets to overcome resistance in BRAFV600E HGG models in vitro and in vivo., Results: We found that, despite causing no major genetic and epigenetic changes, BRAF and/or MEK inhibitor treatment deregulated multiple negative feedback mechanisms, which led to the reactivation of the MAPK pathway through c-Raf and AKT signaling. This altered oncogenic signaling primarily mediated resistance to molecular targeted therapy in BRAFV600E-mutant HGG. To overcome this resistance mechanism, we performed a high-throughput drug screening to identify therapeutic agents that potently induce additive cytotoxicity with BRAF and MEK inhibitors. We discovered that HSP90 inhibition combined with BRAF/MEK inhibition coordinately deactivated the MAPK and AKT/mTOR pathways, and subsequently induced apoptosis via dephosphorylation of GSK3β (Ser9) and inhibition of Bcl-2 family proteins. This mediated potent cytotoxicity in vitro and in vivo in refractory models with acquired resistance to molecular targeted therapy., Conclusions: The combination of an HSP90 inhibitor with BRAF or MEK inhibitors can overcome the limitations of the current therapeutic strategies for BRAFV600E-mutant HGG., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
9. Local Targeting of NAD + Salvage Pathway Alters the Immune Tumor Microenvironment and Enhances Checkpoint Immunotherapy in Glioblastoma.
- Author
-
Li M, Kirtane AR, Kiyokawa J, Nagashima H, Lopes A, Tirmizi ZA, Lee CK, Traverso G, Cahill DP, and Wakimoto H
- Published
- 2021
- Full Text
- View/download PDF
10. TERT Promoter Mutation Analysis for Blood-Based Diagnosis and Monitoring of Gliomas.
- Author
-
Muralidharan K, Yekula A, Small JL, Rosh ZS, Kang KM, Wang L, Lau S, Zhang H, Lee H, Bettegowda C, Chicoine MR, Kalkanis SN, Shankar GM, Nahed BV, Curry WT, Jones PS, Cahill DP, Balaj L, and Carter BS
- Subjects
- Adult, Aged, Brain Neoplasms blood, Brain Neoplasms therapy, Cell Line, Tumor, Cohort Studies, DNA Mutational Analysis methods, Feasibility Studies, Female, Glioma blood, Glioma therapy, Humans, Liquid Biopsy methods, Male, Middle Aged, Mutation, Polymerase Chain Reaction, Promoter Regions, Genetic, Sensitivity and Specificity, Biomarkers, Tumor genetics, Brain Neoplasms diagnosis, Glioma diagnosis, Telomerase genetics
- Abstract
Purpose: Liquid biopsy offers a minimally invasive tool to diagnose and monitor the heterogeneous molecular landscape of tumors over time and therapy. Detection of TERT promoter mutations ( C228T , C250T ) in cfDNA has been successful for some systemic cancers but has yet to be demonstrated in gliomas, despite the high prevalence of these mutations in glioma tissue (>60% of all tumors)., Experimental Design: Here, we developed a novel digital droplet PCR (ddPCR) assay that incorporates features to improve sensitivity and allows for the simultaneous detection and longitudinal monitoring of two TERT promoter mutations ( C228T and C250T ) in cfDNA from the plasma of patients with glioma., Results: In baseline performance in tumor tissue, the assay had perfect concordance with an independently performed clinical pathology laboratory assessment of TERT promoter mutations in the same tumor samples [95% confidence interval (CI), 94%-100%]. Extending to matched plasma samples, we detected TERT mutations in both discovery and blinded multi-institution validation cohorts with an overall sensitivity of 62.5% (95% CI, 52%-73%) and a specificity of 90% (95% CI, 80%-96%) compared with the gold-standard tumor tissue-based detection of TERT mutations. Upon longitudinal monitoring in 5 patients, we report that peripheral TERT -mutant allele frequency reflects the clinical course of the disease, with levels decreasing after surgical intervention and therapy and increasing with tumor progression., Conclusions: Our results demonstrate the feasibility of detecting circulating cfDNA TERT promoter mutations in patients with glioma with clinically relevant sensitivity and specificity., (©2020 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
11. A Hyperactive RelA/p65-Hexokinase 2 Signaling Axis Drives Primary Central Nervous System Lymphoma.
- Author
-
Tateishi K, Miyake Y, Kawazu M, Sasaki N, Nakamura T, Sasame J, Yoshii Y, Ueno T, Miyake A, Watanabe J, Matsushita Y, Shiba N, Udaka N, Ohki K, Fink AL, Tummala SS, Natsumeda M, Ikegaya N, Nishi M, Ohtake M, Miyazaki R, Suenaga J, Murata H, Aoki I, Miller JJ, Fujii Y, Ryo A, Yamanaka S, Mano H, Cahill DP, Wakimoto H, Chi AS, Batchelor TT, Nagane M, Ichimura K, and Yamamoto T
- Subjects
- Animals, CD79 Antigens genetics, Central Nervous System Neoplasms drug therapy, Central Nervous System Neoplasms metabolism, Central Nervous System Neoplasms mortality, Female, Glycolysis, Hexokinase genetics, Humans, Lymphoma drug therapy, Lymphoma metabolism, Lymphoma mortality, Mice, SCID, Mutation, Myeloid Differentiation Factor 88 genetics, NF-kappa B metabolism, NIMA-Interacting Peptidylprolyl Isomerase metabolism, Signal Transduction, Viral Matrix Proteins genetics, Viral Matrix Proteins metabolism, Xenograft Model Antitumor Assays, Central Nervous System Neoplasms pathology, Hexokinase metabolism, Lymphoma pathology, Transcription Factor RelA metabolism
- Abstract
Primary central nervous system lymphoma (PCNSL) is an isolated type of lymphoma of the central nervous system and has a dismal prognosis despite intensive chemotherapy. Recent genomic analyses have identified highly recurrent mutations of MYD88 and CD79B in immunocompetent PCNSL, whereas LMP1 activation is commonly observed in Epstein-Barr virus (EBV)-positive PCNSL. However, a lack of clinically representative preclinical models has hampered our understanding of the pathogenic mechanisms by which genetic aberrations drive PCNSL disease phenotypes. Here, we establish a panel of 12 orthotopic, patient-derived xenograft (PDX) models from both immunocompetent and EBV-positive PCNSL and secondary CNSL biopsy specimens. PDXs faithfully retained their phenotypic, metabolic, and genetic features, with 100% concordance of MYD88 and CD79B mutations present in PCNSL in immunocompetent patients. These models revealed a convergent functional dependency upon a deregulated RelA/p65-hexokinase 2 signaling axis, codriven by either mutated MYD88/CD79B or LMP1 with Pin1 overactivation in immunocompetent PCNSL and EBV-positive PCNSL, respectively. Notably, distinct molecular alterations used by immunocompetent and EBV-positive PCNSL converged to deregulate RelA/p65 expression and to drive glycolysis, which is critical for intracerebral tumor progression and FDG-PET imaging characteristics. Genetic and pharmacologic inhibition of this key signaling axis potently suppressed PCNSL growth in vitro and in vivo . These patient-derived models offer a platform for predicting clinical chemotherapeutics efficacy and provide critical insights into PCNSL pathogenic mechanisms, accelerating therapeutic discovery for this aggressive disease. SIGNIFICANCE: A set of clinically relevant CNSL xenografts identifies a hyperactive RelA/p65-hexokinase 2 signaling axis as a driver of progression and potential therapeutic target for treatment and provides a foundational preclinical platform. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/23/5330/F1.large.jpg., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
12. Local Targeting of NAD + Salvage Pathway Alters the Immune Tumor Microenvironment and Enhances Checkpoint Immunotherapy in Glioblastoma.
- Author
-
Li M, Kirtane AR, Kiyokawa J, Nagashima H, Lopes A, Tirmizi ZA, Lee CK, Traverso G, Cahill DP, and Wakimoto H
- Subjects
- Acrylamides administration & dosage, Animals, Autophagy, B7-H1 Antigen antagonists & inhibitors, B7-H1 Antigen genetics, Brain Neoplasms immunology, Brain Neoplasms metabolism, Brain Neoplasms mortality, CD4-Positive T-Lymphocytes, CD8-Positive T-Lymphocytes, Cell Movement, Cyanides adverse effects, Delayed-Action Preparations, Drug Carriers chemical synthesis, Glioblastoma immunology, Glioblastoma metabolism, Glioblastoma mortality, Guanidines adverse effects, Humans, Injections, Intralesional, Macrophages drug effects, Membrane Proteins metabolism, Mice, NAD analysis, NAD deficiency, Piperidines administration & dosage, Polymers chemical synthesis, RNA, Messenger metabolism, Signal Transduction, Tumor Microenvironment immunology, Up-Regulation drug effects, B7-H1 Antigen metabolism, Brain Neoplasms therapy, Cyanides administration & dosage, Cytokines antagonists & inhibitors, Glioblastoma therapy, Guanidines administration & dosage, NAD drug effects, Nicotinamide Phosphoribosyltransferase antagonists & inhibitors, Tumor Microenvironment drug effects
- Abstract
The aggressive primary brain tumor glioblastoma (GBM) is characterized by aberrant metabolism that fuels its malignant phenotype. Diverse genetic subtypes of malignant glioma are sensitive to selective inhibition of the NAD
+ salvage pathway enzyme nicotinamide phosphoribosyltransferase (NAMPT). However, the potential impact of NAD+ depletion on the brain tumor microenvironment has not been elaborated. In addition, systemic toxicity of NAMPT inhibition remains a significant concern. Here we show that microparticle-mediated intratumoral delivery of NAMPT inhibitor GMX1778 induces specific immunologic changes in the tumor microenvironment of murine GBM, characterized by upregulation of immune checkpoint PD-L1, recruitment of CD3+ , CD4+ , and CD8+ T cells, and reduction of M2-polarized immunosuppressive macrophages. NAD+ depletion and autophagy induced by NAMPT inhibitors mediated the upregulation of PD-L1 transcripts and cell surface protein levels in GBM cells. NAMPT inhibitor modulation of the tumor immune microenvironment was therefore combined with PD-1 checkpoint blockade in vivo , significantly increasing the survival of GBM-bearing animals. Thus, the therapeutic impacts of NAMPT inhibition extended beyond neoplastic cells, shaping surrounding immune effectors. Microparticle delivery and release of NAMPT inhibitor at the tumor site offers a safe and robust means to alter an immune tumor microenvironment that could potentiate checkpoint immunotherapy for glioblastoma. SIGNIFICANCE: Microparticle-mediated local inhibition of NAMPT modulates the tumor immune microenvironment and acts cooperatively with anti-PD-1 checkpoint blockade, offering a combination immunotherapy strategy for the treatment of GBM., (©2020 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
13. Poly(ADP-ribose) Glycohydrolase Inhibition Sequesters NAD + to Potentiate the Metabolic Lethality of Alkylating Chemotherapy in IDH-Mutant Tumor Cells.
- Author
-
Nagashima H, Lee CK, Tateishi K, Higuchi F, Subramanian M, Rafferty S, Melamed L, Miller JJ, Wakimoto H, and Cahill DP
- Subjects
- Humans, Tumor Cells, Cultured, Antineoplastic Agents, Alkylating therapeutic use, Glycoside Hydrolases metabolism, NAD metabolism
- Abstract
NAD
+ is an essential cofactor metabolite and is the currency of metabolic transactions critical for cell survival. Depending on tissue context and genotype, cancer cells have unique dependencies on NAD+ metabolic pathways. PARPs catalyze oligomerization of NAD+ monomers into PAR chains during cellular response to alkylating chemotherapeutics, including procarbazine or temozolomide. Here we find that, in endogenous IDH1-mutant tumor models, alkylator-induced cytotoxicity is markedly augmented by pharmacologic inhibition or genetic knockout of the PAR breakdown enzyme PAR glycohydrolase (PARG). Both in vitro and in vivo , we observe that concurrent alkylator and PARG inhibition depletes freely available NAD+ by preventing PAR breakdown, resulting in NAD+ sequestration and collapse of metabolic homeostasis. This effect reversed with NAD+ rescue supplementation, confirming the mechanistic basis of cytotoxicity. Thus, alkylating chemotherapy exposes a genotype-specific metabolic weakness in tumor cells that can be exploited by PARG inactivation. SIGNIFICANCE: Oncogenic mutations in the isocitrate dehydrogenase genes IDH1 or IDH2 initiate diffuse gliomas of younger adulthood. Strategies to maximize the effectiveness of chemotherapy in these tumors are needed. We discover alkylating chemotherapy and concurrent PARG inhibition exploits an intrinsic metabolic weakness within these cancer cells to provide genotype-specific benefit. See related commentary by Pirozzi and Yan, p. 1629 . This article is highlighted in the In This Issue feature, p. 1611 ., (©2020 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
14. Restoration of Temozolomide Sensitivity by PARP Inhibitors in Mismatch Repair Deficient Glioblastoma is Independent of Base Excision Repair.
- Author
-
Higuchi F, Nagashima H, Ning J, Koerner MVA, Wakimoto H, and Cahill DP
- Subjects
- Animals, Antineoplastic Agents, Alkylating pharmacology, Cell Line, Tumor, Female, Glioblastoma genetics, Glioblastoma metabolism, Glioblastoma pathology, Humans, Mice, Xenograft Model Antitumor Assays, DNA Mismatch Repair, DNA Repair, Drug Resistance, Neoplasm, Glioblastoma drug therapy, Phthalazines pharmacology, Piperazines pharmacology, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Temozolomide pharmacology
- Abstract
Purpose: Emergence of mismatch repair (MMR) deficiency is a frequent mechanism of acquired resistance to the alkylating chemotherapeutic temozolomide (TMZ) in gliomas. Poly(ADP-ribose) polymerase inhibitors (PARPi) have been shown to potentiate TMZ cytotoxicity in several cancer types, including gliomas. We tested whether PARP inhibition could re-sensitize MSH6-null MMR-deficient gliomas to TMZ, and assessed the role of the base excision repair (BER) DNA damage repair pathway in PARPi-mediated effects., Experimental Design: Isogenic pairs of MSH6 wild-type and MSH6-inactivated human glioblastoma (GBM) cells (including both IDH1/2 wild-type and IDH1 mutant), as well as MSH6-null cells derived from a patient with recurrent GBM were treated with TMZ, the PARPi veliparib or olaparib, and combination thereof. Efficacy of PARPi combined with TMZ was assessed in vivo . We used genetic and pharmacological approaches to dissect the contribution of BER., Results: While having no detectable effect in MSH6 wild-type GBMs, PARPi selectively restored TMZ sensitivity in MSH6-deficient GBM cells. This genotype-specific restoration of activity translated in vivo , where combination treatment of veliparib and TMZ showed potent suppression of tumor growth of MSH6-inactivated orthotopic xenografts, compared with TMZ monotherapy. Unlike PARPi, genetic and pharmacological blockage of BER pathway did not re-sensitize MSH6-inactivated GBM cells to TMZ. Similarly, CRISPR PARP1 knockout did not re-sensitize MSH6-inactivated GBM cells to TMZ., Conclusions: PARPi restoration of TMZ chemosensitivity in MSH6-inactivated glioma represents a promising strategy to overcome acquired chemoresistance caused by MMR deficiency. Mechanistically, this PARPi-mediated synthetic phenotype was independent of BER blockage and was not recapitulated by loss of PARP1., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
15. PI3K/AKT/mTOR Pathway Alterations Promote Malignant Progression and Xenograft Formation in Oligodendroglial Tumors.
- Author
-
Tateishi K, Nakamura T, Juratli TA, Williams EA, Matsushita Y, Miyake S, Nishi M, Miller JJ, Tummala SS, Fink AL, Lelic N, Koerner MVA, Miyake Y, Sasame J, Fujimoto K, Tanaka T, Minamimoto R, Matsunaga S, Mukaihara S, Shuto T, Taguchi H, Udaka N, Murata H, Ryo A, Yamanaka S, Curry WT, Dias-Santagata D, Yamamoto T, Ichimura K, Batchelor TT, Chi AS, Iafrate AJ, Wakimoto H, and Cahill DP
- Subjects
- Animals, Brain Neoplasms drug therapy, Brain Neoplasms metabolism, Cell Line, Tumor, Class I Phosphatidylinositol 3-Kinases antagonists & inhibitors, Class I Phosphatidylinositol 3-Kinases genetics, Female, Humans, Mice, Mice, SCID, Oligodendroglioma drug therapy, Oligodendroglioma metabolism, Proto-Oncogene Proteins c-akt antagonists & inhibitors, Signal Transduction drug effects, TOR Serine-Threonine Kinases antagonists & inhibitors, Xenograft Model Antitumor Assays, Brain Neoplasms pathology, Class I Phosphatidylinositol 3-Kinases metabolism, Oligodendroglioma pathology, Phosphoinositide-3 Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-akt metabolism, TOR Serine-Threonine Kinases metabolism
- Abstract
Purpose: Oligodendroglioma has a relatively favorable prognosis, however, often undergoes malignant progression. We hypothesized that preclinical models of oligodendroglioma could facilitate identification of therapeutic targets in progressive oligodendroglioma. We established multiple oligodendroglioma xenografts to determine if the PI3K/AKT/mTOR signaling pathway drives tumor progression., Experimental Design: Two anatomically distinct tumor samples from a patient who developed progressive anaplastic oligodendroglioma (AOD) were collected for orthotopic transplantation in mice. We additionally implanted 13 tumors to investigate the relationship between PI3K/AKT/mTOR pathway alterations and oligodendroglioma xenograft formation. Pharmacologic vulnerabilities were tested in newly developed AOD models in vitro and in vivo ., Results: A specimen from the tumor site that subsequently manifested rapid clinical progression contained a PIK3CA mutation E542K, and yielded propagating xenografts that retained the OD/AOD-defining genomic alterations ( IDH1
R132H and 1p/19q codeletion) and PIK3CAE542K , and displayed characteristic sensitivity to alkylating chemotherapeutic agents. In contrast, a xenograft did not engraft from the region that was clinically stable and had wild-type PIK3CA . In our panel of OD/AOD xenografts, the presence of activating mutations in the PI3K/AKT/mTOR pathway was consistently associated with xenograft establishment (6/6, 100%). OD/AOD that failed to generate xenografts did not have activating PI3K/AKT/mTOR alterations (0/9, P < 0.0001). Importantly, mutant PIK3CA oligodendroglioma xenografts were vulnerable to PI3K/AKT/mTOR pathway inhibitors in vitro and in vivo -evidence that mutant PIK3CA is a tumorigenic driver in oligodendroglioma., Conclusions: Activation of the PI3K/AKT/mTOR pathway is an oncogenic driver and is associated with xenograft formation in oligodendrogliomas. These findings have implications for therapeutic targeting of PI3K/AKT/mTOR pathway activation in progressive oligodendrogliomas., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
16. The Dual PI3K/mTOR Pathway Inhibitor GDC-0084 Achieves Antitumor Activity in PIK3CA -Mutant Breast Cancer Brain Metastases.
- Author
-
Ippen FM, Alvarez-Breckenridge CA, Kuter BM, Fink AL, Bihun IV, Lastrapes M, Penson T, Schmidt SP, Wojtkiewicz GR, Ning J, Subramanian M, Giobbie-Hurder A, Martinez-Lage M, Carter SL, Cahill DP, Wakimoto H, and Brastianos PK
- Subjects
- Animals, Antineoplastic Agents pharmacology, Apoptosis drug effects, Brain Neoplasms drug therapy, Breast Neoplasms diagnostic imaging, Breast Neoplasms metabolism, Cell Line, Tumor, Cell Proliferation drug effects, Disease Models, Animal, Female, Humans, Immunohistochemistry, Mice, Protein Kinase Inhibitors pharmacology, Brain Neoplasms metabolism, Brain Neoplasms secondary, Breast Neoplasms genetics, Breast Neoplasms pathology, Class I Phosphatidylinositol 3-Kinases genetics, Class I Phosphatidylinositol 3-Kinases metabolism, Oxazines pharmacology, Pyrimidines pharmacology, Signal Transduction drug effects, TOR Serine-Threonine Kinases metabolism
- Abstract
Purpose: Previous studies have shown that the PI3K/Akt/mTOR pathway is activated in up to 70% of breast cancer brain metastases, but there are no approved agents for affected patients. GDC-0084 is a brain penetrant, dual PI3K/mTOR inhibitor that has shown promising activity in a preclinical model of glioblastoma. The aim of this study was to analyze the efficacy of PI3K/mTOR blockade in breast cancer brain metastases models. Experimental Design: The efficacy of GDC-0084 was evaluated in PIK3CA -mutant and PIK3CA wild-type breast cancer cell lines and the isogenic pairs of PIK3CA wild-type and mutant (H1047R/+) MCF10A cells in vitro . In vitro studies included cell viability and apoptosis assays, cell-cycle analysis, and Western blots. In vivo , the effect of GDC-0084 was investigated in breast cancer brain metastasis xenograft mouse models and assessed by bioluminescent imaging and IHC., Results: In vitro , GDC-0084 considerably decreased cell viability, induced apoptosis, and inhibited phosphorylation of Akt and p70 S6 kinase in a dose-dependent manner in PIK3CA -mutant breast cancer brain metastatic cell lines. In contrast, GDC-0084 led only to growth inhibition in PIK3CA wild-type cell lines in vitro . In vivo , treatment with GDC-0084 markedly inhibited the growth of PIK3CA -mutant, with accompanying signaling changes, and not PIK3CA wild-type brain tumors., Conclusions: The results of this study suggest that the brain-penetrant PI3K/mTOR targeting GDC-0084 is a promising treatment option for breast cancer brain metastases with dysregulated PI3K/mTOR signaling pathway conferred by activating PIK3CA mutations. A national clinical trial is planned to further investigate the role of this compound in patients with brain metastases., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
17. Cell Surface Notch Ligand DLL3 is a Therapeutic Target in Isocitrate Dehydrogenase-mutant Glioma.
- Author
-
Spino M, Kurz SC, Chiriboga L, Serrano J, Zeck B, Sen N, Patel S, Shen G, Vasudevaraja V, Tsirigos A, Suryadevara CM, Frenster JD, Tateishi K, Wakimoto H, Jain R, Riina HA, Nicolaides TP, Sulman EP, Cahill DP, Golfinos JG, Isse K, Saunders LR, Zagzag D, Placantonakis DG, Snuderl M, and Chi AS
- Subjects
- Antibodies, Monoclonal, Humanized genetics, Antibodies, Monoclonal, Humanized therapeutic use, Benzodiazepinones therapeutic use, Brain pathology, DNA Methylation genetics, Female, Gene Expression Regulation, Neoplastic drug effects, Genotype, Glioma genetics, Glioma pathology, Humans, Immunoconjugates genetics, Immunoconjugates therapeutic use, Ligands, Male, Mutation, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local genetics, Neoplasm Recurrence, Local pathology, RNA genetics, Receptors, Notch genetics, Glioma drug therapy, Intracellular Signaling Peptides and Proteins genetics, Isocitrate Dehydrogenase genetics, Membrane Proteins genetics, Molecular Targeted Therapy
- Abstract
Purpose: Isocitrate dehydrogenase ( IDH )-mutant glioma is a distinct glioma molecular subtype for which no effective molecularly directed therapy exists. Low-grade gliomas, which are 80%-90% IDH -mutant, have high RNA levels of the cell surface Notch ligand DLL3. We sought to determine DLL3 expression by IHC in glioma molecular subtypes and the potential efficacy of an anti-DLL3 antibody-drug conjugate (ADC), rovalpituzumab tesirine (Rova-T), in IDH -mutant glioma., Experimental Design: We evaluated DLL3 expression by RNA using TCGA data and by IHC in a discovery set of 63 gliomas and 20 nontumor brain tissues and a validation set of 62 known IDH wild-type and mutant gliomas using a monoclonal anti-DLL3 antibody. Genotype was determined using a DNA methylation array classifier or by sequencing. The effect of Rova-T on patient-derived endogenous IDH -mutant glioma tumorspheres was determined by cell viability assay., Results: Compared to IDH wild-type glioblastoma, IDH -mutant gliomas have significantly higher DLL3 RNA ( P < 1 × 10
-15 ) and protein by IHC ( P = 0.0014 and P < 4.3 × 10-6 in the discovery and validation set, respectively). DLL3 immunostaining was intense and homogeneous in IDH -mutant gliomas, retained in all recurrent tumors, and detected in only 1 of 20 nontumor brains. Patient-derived IDH -mutant glioma tumorspheres overexpressed DLL3 and were potently sensitive to Rova-T in an antigen-dependent manner., Conclusions: DLL3 is selectively and homogeneously expressed in IDH -mutant gliomas and can be targeted with Rova-T in patient-derived IDH -mutant glioma tumorspheres. Our findings are potentially immediately translatable and have implications for therapeutic strategies that exploit cell surface tumor-associated antigens., (©2018 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
18. Exploiting MCL1 Dependency with Combination MEK + MCL1 Inhibitors Leads to Induction of Apoptosis and Tumor Regression in KRAS -Mutant Non-Small Cell Lung Cancer.
- Author
-
Nangia V, Siddiqui FM, Caenepeel S, Timonina D, Bilton SJ, Phan N, Gomez-Caraballo M, Archibald HL, Li C, Fraser C, Rigas D, Vajda K, Ferris LA, Lanuti M, Wright CD, Raskin KA, Cahill DP, Shin JH, Keyes C, Sequist LV, Piotrowska Z, Farago AF, Azzoli CG, Gainor JF, Sarosiek KA, Brown SP, Coxon A, Benes CH, Hughes PE, and Hata AN
- Subjects
- A549 Cells, Aniline Compounds administration & dosage, Aniline Compounds pharmacology, Animals, Antineoplastic Agents administration & dosage, Antineoplastic Agents pharmacology, Apoptosis drug effects, Benzamides pharmacology, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung metabolism, Cell Line, Tumor, Diphenylamine analogs & derivatives, Diphenylamine pharmacology, Humans, Lung Neoplasms genetics, Lung Neoplasms metabolism, Mice, Knockout, Mice, Nude, Mice, SCID, Mitogen-Activated Protein Kinase Kinases metabolism, Myeloid Cell Leukemia Sequence 1 Protein genetics, Myeloid Cell Leukemia Sequence 1 Protein metabolism, Protein Kinase Inhibitors administration & dosage, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) metabolism, Sulfonamides administration & dosage, Sulfonamides pharmacology, Tumor Burden drug effects, Xenograft Model Antitumor Assays methods, Antineoplastic Combined Chemotherapy Protocols pharmacology, Carcinoma, Non-Small-Cell Lung drug therapy, Lung Neoplasms drug therapy, Mitogen-Activated Protein Kinase Kinases antagonists & inhibitors, Myeloid Cell Leukemia Sequence 1 Protein antagonists & inhibitors, Proto-Oncogene Proteins p21(ras) antagonists & inhibitors
- Abstract
BH3 mimetic drugs, which inhibit prosurvival BCL2 family proteins, have limited single-agent activity in solid tumor models. The potential of BH3 mimetics for these cancers may depend on their ability to potentiate the apoptotic response to chemotherapy and targeted therapies. Using a novel class of potent and selective MCL1 inhibitors, we demonstrate that concurrent MEK + MCL1 inhibition induces apoptosis and tumor regression in KRAS -mutant non-small cell lung cancer (NSCLC) models, which respond poorly to MEK inhibition alone. Susceptibility to BH3 mimetics that target either MCL1 or BCL-xL was determined by the differential binding of proapoptotic BCL2 proteins to MCL1 or BCL-xL, respectively. The efficacy of dual MEK + MCL1 blockade was augmented by prior transient exposure to BCL-xL inhibitors, which promotes the binding of proapoptotic BCL2 proteins to MCL1. This suggests a novel strategy for integrating BH3 mimetics that target different BCL2 family proteins for KRAS -mutant NSCLC. SIGNIFICANCE: Defining the molecular basis for MCL1 versus BCL-xL dependency will be essential for effective prioritization of BH3 mimetic combination therapies in the clinic. We discover a novel strategy for integrating BCL-xL and MCL1 inhibitors to drive and subsequently exploit apoptotic dependencies of KRAS -mutant NSCLCs treated with MEK inhibitors. See related commentary by Leber et al., p. 1511 . This article is highlighted in the In This Issue feature, p. 1494 ., (©2018 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
19. The Alkylating Chemotherapeutic Temozolomide Induces Metabolic Stress in IDH1 -Mutant Cancers and Potentiates NAD + Depletion-Mediated Cytotoxicity.
- Author
-
Tateishi K, Higuchi F, Miller JJ, Koerner MVA, Lelic N, Shankar GM, Tanaka S, Fisher DE, Batchelor TT, Iafrate AJ, Wakimoto H, Chi AS, and Cahill DP
- Subjects
- Acrylamides pharmacology, Animals, Cell Line, Tumor, Dacarbazine pharmacology, Enzyme Inhibitors pharmacology, Female, Glioma genetics, Humans, Isocitrate Dehydrogenase genetics, Mice, Mice, SCID, Mutation, NAD, Nicotinamide Phosphoribosyltransferase antagonists & inhibitors, Piperidines pharmacology, Random Allocation, Temozolomide, Xenograft Model Antitumor Assays, Antineoplastic Agents, Alkylating pharmacology, Dacarbazine analogs & derivatives, Glioma metabolism, Stress, Physiological drug effects
- Abstract
IDH1 -mutant gliomas are dependent upon the canonical coenzyme NAD
+ for survival. It is known that PARP activation consumes NAD+ during base excision repair (BER) of chemotherapy-induced DNA damage. We therefore hypothesized that a strategy combining NAD+ biosynthesis inhibitors with the alkylating chemotherapeutic agent temozolomide could potentiate NAD+ depletion-mediated cytotoxicity in mutant IDH1 cancer cells. To investigate the impact of temozolomide on NAD+ metabolism, patient-derived xenografts and engineered mutant IDH1 -expressing cell lines were exposed to temozolomide, in vitro and in vivo , both alone and in combination with nicotinamide phosphoribosyltransferase (NAMPT) inhibitors, which block NAD+ biosynthesis. The acute time period (<3 hours) after temozolomide treatment displayed a burst of NAD+ consumption driven by PARP activation. In IDH1 -mutant-expressing cells, this consumption reduced further the abnormally lowered basal steady-state levels of NAD+ , introducing a window of hypervulnerability to NAD+ biosynthesis inhibitors. This effect was selective for IDH1 -mutant cells and independent of methylguanine methyltransferase or mismatch repair status, which are known rate-limiting mediators of adjuvant temozolomide genotoxic sensitivity. Combined temozolomide and NAMPT inhibition in an in vivo IDH1 -mutant cancer model exhibited enhanced efficacy compared with each agent alone. Thus, we find IDH1 -mutant cancers have distinct metabolic stress responses to chemotherapy-induced DNA damage and that combination regimens targeting nonredundant NAD+ pathways yield potent anticancer efficacy in vivo Such targeting of convergent metabolic pathways in genetically selected cancers could minimize treatment toxicity and improve durability of response to therapy. Cancer Res; 77(15); 4102-15. ©2017 AACR ., (©2017 American Association for Cancer Research.)- Published
- 2017
- Full Text
- View/download PDF
20. Spatial Proximity to Fibroblasts Impacts Molecular Features and Therapeutic Sensitivity of Breast Cancer Cells Influencing Clinical Outcomes.
- Author
-
Marusyk A, Tabassum DP, Janiszewska M, Place AE, Trinh A, Rozhok AI, Pyne S, Guerriero JL, Shu S, Ekram M, Ishkin A, Cahill DP, Nikolsky Y, Chan TA, Rimawi MF, Hilsenbeck S, Schiff R, Osborne KC, Letai A, and Polyak K
- Subjects
- Breast Neoplasms pathology, Cell Line, Tumor, Humans, Treatment Outcome, Breast Neoplasms metabolism, Fibroblasts metabolism, Gene Expression Profiling methods
- Abstract
Using a three-dimensional coculture model, we identified significant subtype-specific changes in gene expression, metabolic, and therapeutic sensitivity profiles of breast cancer cells in contact with cancer-associated fibroblasts (CAF). CAF-induced gene expression signatures predicted clinical outcome and immune-related differences in the microenvironment. We found that fibroblasts strongly protect carcinoma cells from lapatinib, attributable to its reduced accumulation in carcinoma cells and an elevated apoptotic threshold. Fibroblasts from normal breast tissues and stromal cultures of brain metastases of breast cancer had similar effects as CAFs. Using synthetic lethality approaches, we identified molecular pathways whose inhibition sensitizes HER2
+ breast cancer cells to lapatinib both in vitro and in vivo, including JAK2/STAT3 and hyaluronic acid. Neoadjuvant lapatinib therapy in HER2+ breast tumors lead to a significant increase of phospho-STAT3+ cancer cells and a decrease in the spatial proximity of proliferating (Ki67+ ) cells to CAFs impacting therapeutic responses. Our studies identify CAF-induced physiologically and clinically relevant changes in cancer cells and offer novel approaches for overcoming microenvironment-mediated therapeutic resistance. Cancer Res; 76(22); 6495-506. ©2016 AACR., (©2016 American Association for Cancer Research.)- Published
- 2016
- Full Text
- View/download PDF
21. Myc-Driven Glycolysis Is a Therapeutic Target in Glioblastoma.
- Author
-
Tateishi K, Iafrate AJ, Ho Q, Curry WT, Batchelor TT, Flaherty KT, Onozato ML, Lelic N, Sundaram S, Cahill DP, Chi AS, and Wakimoto H
- Subjects
- Animals, Apoptosis genetics, Cell Line, Tumor, Disease Models, Animal, Gene Amplification, Glioblastoma drug therapy, Glycolysis genetics, Humans, Mice, NAD metabolism, Nicotinamide Phosphoribosyltransferase antagonists & inhibitors, Nicotinamide Phosphoribosyltransferase metabolism, Proto-Oncogene Proteins c-myc metabolism, RNA Interference, RNA, Small Interfering genetics, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Glioblastoma genetics, Glioblastoma metabolism, Glucose metabolism, Proto-Oncogene Proteins c-myc genetics
- Abstract
Purpose: Deregulated Myc drives an oncogenic metabolic state, including pseudohypoxic glycolysis, adapted for the constitutive production of biomolecular precursors to feed rapid tumor cell growth. In glioblastoma, Myc facilitates renewal of the tumor-initiating cell reservoir contributing to tumor maintenance. We investigated whether targeting the Myc-driven metabolic state could be a selectively toxic therapeutic strategy for glioblastoma., Experimental Design: The glycolytic dependency of Myc-driven glioblastoma was tested using (13)C metabolic flux analysis, glucose-limiting culture assays, and glycolysis inhibitors, including inhibitors of the NAD(+) salvage enzyme nicotinamide phosphoribosyl-transferase (NAMPT), in MYC and MYCN shRNA knockdown and lentivirus overexpression systems and in patient-derived glioblastoma tumorspheres with and without MYC/MYCN amplification. The in vivo efficacy of glycolyic inhibition was tested using NAMPT inhibitors in MYCN-amplified patient-derived glioblastoma orthotopic xenograft mouse models., Results: Enforced Myc overexpression increased glucose flux and expression of glycolytic enzymes in glioblastoma cells. Myc and N-Myc knockdown and Myc overexpression systems demonstrated that Myc activity determined sensitivity and resistance to inhibition of glycolysis. Small-molecule inhibitors of glycolysis, particularly NAMPT inhibitors, were selectively toxic to MYC/MYCN-amplified patient-derived glioblastoma tumorspheres. NAMPT inhibitors were potently cytotoxic, inducing apoptosis and significantly extended the survival of mice bearing MYCN-amplified patient-derived glioblastoma orthotopic xenografts., Conclusions: Myc activation in glioblastoma generates a dependency on glycolysis and an addiction to metabolites required for glycolysis. Glycolytic inhibition via NAMPT inhibition represents a novel metabolically targeted therapeutic strategy for MYC or MYCN-amplified glioblastoma and potentially other cancers genetically driven by Myc. Clin Cancer Res; 22(17); 4452-65. ©2016 AACR., Competing Interests: All authors have no conflicts of interest to report with regard to this manuscript., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
22. Treatment Response Assessment in IDH-Mutant Glioma Patients by Noninvasive 3D Functional Spectroscopic Mapping of 2-Hydroxyglutarate.
- Author
-
Andronesi OC, Loebel F, Bogner W, Marjańska M, Vander Heiden MG, Iafrate AJ, Dietrich J, Batchelor TT, Gerstner ER, Kaelin WG, Chi AS, Rosen BR, and Cahill DP
- Subjects
- Biomarkers, Brain pathology, Brain Neoplasms diagnosis, Brain Neoplasms genetics, Brain Neoplasms metabolism, Brain Neoplasms therapy, Chemoradiotherapy, Adjuvant, Female, Glioma therapy, Humans, Image Processing, Computer-Assisted, Imaging, Three-Dimensional, Male, Neoplasm Grading, Neoplasm Staging, Glioma diagnosis, Glioma genetics, Glioma metabolism, Glutarates metabolism, Isocitrate Dehydrogenase genetics, Isocitrate Dehydrogenase metabolism, Magnetic Resonance Spectroscopy methods, Mutation
- Abstract
Purpose: Measurements of objective response rates are critical to evaluate new glioma therapies. The hallmark metabolic alteration in gliomas with mutant isocitrate dehydrogenase (IDH) is the overproduction of oncometabolite 2-hydroxyglutarate (2HG), which plays a key role in malignant transformation. 2HG represents an ideal biomarker to probe treatment response in IDH-mutant glioma patients, and we hypothesized a decrease in 2HG levels would be measureable by in vivo magnetic resonance spectroscopy (MRS) as a result of antitumor therapy., Experimental Design: We report a prospective longitudinal imaging study performed in 25 IDH-mutant glioma patients receiving adjuvant radiation and chemotherapy. A newly developed 3D MRS imaging was used to noninvasively image 2HG. Paired Student t test was used to compare pre- and posttreatment tumor 2HG values. Test-retest measurements were performed to determine the threshold for 2HG functional spectroscopic maps (fSM). Univariate and multivariate regression were performed to correlate 2HG changes with Karnofsky performance score (KPS)., Results: We found that mean 2HG (2HG/Cre) levels decreased significantly (median = 48.1%; 95% confidence interval = 27.3%-56.5%;P= 0.007) in the posttreatment scan. The volume of decreased 2HG correlates (R(2)= 0.88,P= 0.002) with clinical status evaluated by KPS., Conclusions: We demonstrate that dynamic measurements of 2HG are feasible by 3D fSM, and the decrease of 2HG levels can monitor treatment response in patients with IDH-mutant gliomas. Our results indicate that quantitative in vivo 2HG imaging may be used for precision medicine and early response assessment in clinical trials of therapies targeting IDH-mutant gliomas., (©2015 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
23. Genomic Characterization of Brain Metastases Reveals Branched Evolution and Potential Therapeutic Targets.
- Author
-
Brastianos PK, Carter SL, Santagata S, Cahill DP, Taylor-Weiner A, Jones RT, Van Allen EM, Lawrence MS, Horowitz PM, Cibulskis K, Ligon KL, Tabernero J, Seoane J, Martinez-Saez E, Curry WT, Dunn IF, Paek SH, Park SH, McKenna A, Chevalier A, Rosenberg M, Barker FG 2nd, Gill CM, Van Hummelen P, Thorner AR, Johnson BE, Hoang MP, Choueiri TK, Signoretti S, Sougnez C, Rabin MS, Lin NU, Winer EP, Stemmer-Rachamimov A, Meyerson M, Garraway L, Gabriel S, Lander ES, Beroukhim R, Batchelor TT, Baselga J, Louis DN, Getz G, and Hahn WC
- Subjects
- Brain Neoplasms diagnosis, Brain Neoplasms drug therapy, Cluster Analysis, Disease Progression, Drug Resistance, Neoplasm genetics, Exome, Genetic Heterogeneity, Genetic Variation, Genome-Wide Association Study, High-Throughput Nucleotide Sequencing, Humans, Lymph Nodes metabolism, Lymph Nodes pathology, Molecular Targeted Therapy, Mutation, Neoplasms metabolism, Signal Transduction drug effects, Brain Neoplasms genetics, Brain Neoplasms secondary, Neoplasms genetics, Neoplasms pathology
- Abstract
Unlabelled: Brain metastases are associated with a dismal prognosis. Whether brain metastases harbor distinct genetic alterations beyond those observed in primary tumors is unknown. We performed whole-exome sequencing of 86 matched brain metastases, primary tumors, and normal tissue. In all clonally related cancer samples, we observed branched evolution, where all metastatic and primary sites shared a common ancestor yet continued to evolve independently. In 53% of cases, we found potentially clinically informative alterations in the brain metastases not detected in the matched primary-tumor sample. In contrast, spatially and temporally separated brain metastasis sites were genetically homogenous. Distal extracranial and regional lymph node metastases were highly divergent from brain metastases. We detected alterations associated with sensitivity to PI3K/AKT/mTOR, CDK, and HER2/EGFR inhibitors in the brain metastases. Genomic analysis of brain metastases provides an opportunity to identify potentially clinically informative alterations not detected in clinically sampled primary tumors, regional lymph nodes, or extracranial metastases., Significance: Decisions for individualized therapies in patients with brain metastasis are often made from primary-tumor biopsies. We demonstrate that clinically actionable alterations present in brain metastases are frequently not detected in primary biopsies, suggesting that sequencing of primary biopsies alone may miss a substantial number of opportunities for targeted therapy., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
24. Targetable signaling pathway mutations are associated with malignant phenotype in IDH-mutant gliomas.
- Author
-
Wakimoto H, Tanaka S, Curry WT, Loebel F, Zhao D, Tateishi K, Chen J, Klofas LK, Lelic N, Kim JC, Dias-Santagata D, Ellisen LW, Borger DR, Fendt SM, Vander Heiden MG, Batchelor TT, Iafrate AJ, Cahill DP, and Chi AS
- Subjects
- Animals, Brain Neoplasms mortality, Cell Transformation, Neoplastic genetics, Disease-Free Survival, Gas Chromatography-Mass Spectrometry, Glioma mortality, Heterografts, Humans, In Situ Hybridization, Fluorescence, Kaplan-Meier Estimate, Mice, Mutation, Oncogenes, Phenotype, Brain Neoplasms genetics, Brain Neoplasms pathology, Glioma genetics, Glioma pathology, Isocitrate Dehydrogenase genetics
- Abstract
Purpose: Isocitrate dehydrogenase (IDH) gene mutations occur in low-grade and high-grade gliomas. We sought to identify the genetic basis of malignant phenotype heterogeneity in IDH-mutant gliomas., Methods: We prospectively implanted tumor specimens from 20 consecutive IDH1-mutant glioma resections into mouse brains and genotyped all resection specimens using a CLIA-certified molecular panel. Gliomas with cancer driver mutations were tested for sensitivity to targeted inhibitors in vitro. Associations between genomic alterations and outcomes were analyzed in patients., Results: By 10 months, 8 of 20 IDH1-mutant gliomas developed intracerebral xenografts. All xenografts maintained mutant IDH1 and high levels of 2-hydroxyglutarate on serial transplantation. All xenograft-producing gliomas harbored "lineage-defining" mutations in CIC (oligodendroglioma) or TP53 (astrocytoma), and 6 of 8 additionally had activating mutations in PIK3CA or amplification of PDGFRA, MET, or N-MYC. Only IDH1 and CIC/TP53 mutations were detected in non-xenograft-forming gliomas (P = 0.0007). Targeted inhibition of the additional alterations decreased proliferation in vitro. Moreover, we detected alterations in known cancer driver genes in 13.4% of IDH-mutant glioma patients, including PIK3CA, KRAS, AKT, or PTEN mutation or PDGFRA, MET, or N-MYC amplification. IDH/CIC mutant tumors were associated with PIK3CA/KRAS mutations whereas IDH/TP53 tumors correlated with PDGFRA/MET amplification. Presence of driver alterations at progression was associated with shorter subsequent progression-free survival (median 9.0 vs. 36.1 months; P = 0.0011)., Conclusion: A subset of IDH-mutant gliomas with mutations in driver oncogenes has a more malignant phenotype in patients. Identification of these alterations may provide an opportunity for use of targeted therapies in these patients. Clin Cancer Res; 20(11); 2898-909. ©2014 AACR., (©2014 American Association for Cancer Research.)
- Published
- 2014
- Full Text
- View/download PDF
25. MSH6 mutations arise in glioblastomas during temozolomide therapy and mediate temozolomide resistance.
- Author
-
Yip S, Miao J, Cahill DP, Iafrate AJ, Aldape K, Nutt CL, and Louis DN
- Subjects
- Antineoplastic Agents, Alkylating pharmacology, Antineoplastic Agents, Alkylating therapeutic use, Base Sequence, Blotting, Western, Cell Line, Tumor, Cell Survival drug effects, DNA Mutational Analysis, DNA-Binding Proteins metabolism, Dacarbazine pharmacology, Dacarbazine therapeutic use, Dose-Response Relationship, Drug, Drug Resistance, Neoplasm genetics, Glioblastoma drug therapy, Glioblastoma pathology, Humans, Microsatellite Instability, RNA Interference, Temozolomide, Tumor Cells, Cultured, DNA-Binding Proteins genetics, Dacarbazine analogs & derivatives, Glioblastoma genetics, Mutation
- Abstract
Purpose: Over the past few years, the alkylating agent temozolomide has become the standard-of-care therapy for patients with glioblastoma, the most common brain tumor. Recently, large-scale cancer genome sequencing efforts have identified a hypermutation phenotype and inactivating MSH6 mismatch repair gene mutations in recurrent, post-temozolomide glioblastomas, particularly those growing more rapidly during temozolomide treatment. This study aimed to clarify the timing and role of MSH6 mutations in mediating glioblastoma temozolomide resistance., Experimental Design: MSH6 sequence and microsatellite instability (MSI) status were determined in matched prechemotherapy and postchemotherapy glioblastomas identified by The Cancer Genome Atlas (TCGA) as having posttreatment MSH6 mutations. Temozolomide-resistant lines were derived in vitro through selective growth under temozolomide, and the MSH6 gene was sequenced in resistant clones. The role of MSH6 inactivation in mediating resistance was explored using lentiviral short hairpin RNA knockdown and MSH6 reconstitution., Results: MSH6 mutations were confirmed in posttreatment TCGA glioblastomas but absent in matched pretreatment tumors. The posttreatment hypermutation phenotype displayed a signature bias toward CpC transitions and was not associated with MSI. In vitro modeling through exposure of an MSH6 wild-type glioblastoma line to temozolomide resulted in resistant clones; one clone showed an MSH6 mutation, Thr(1219)Ile, that had been independently noted in two treated TCGA glioblastomas. Knockdown of MSH6 in the glioblastoma line U251 increased resistance to temozolomide cytotoxicity and reconstitution restored cytotoxicity in MSH6-null glioma cells., Conclusions: MSH6 mutations are selected in glioblastomas during temozolomide therapy both in vitro and in vivo and are causally associated with temozolomide resistance.
- Published
- 2009
- Full Text
- View/download PDF
26. Loss of the mismatch repair protein MSH6 in human glioblastomas is associated with tumor progression during temozolomide treatment.
- Author
-
Cahill DP, Levine KK, Betensky RA, Codd PJ, Romany CA, Reavie LB, Batchelor TT, Futreal PA, Stratton MR, Curry WT, Iafrate AJ, and Louis DN
- Subjects
- Adult, Aged, Brain Neoplasms drug therapy, Brain Neoplasms pathology, Cell Proliferation drug effects, DNA Modification Methylases biosynthesis, DNA Modification Methylases drug effects, DNA Repair Enzymes biosynthesis, DNA Repair Enzymes drug effects, DNA-Binding Proteins biosynthesis, Dacarbazine adverse effects, Disease Progression, Female, Glioblastoma drug therapy, Glioblastoma pathology, Humans, Immunohistochemistry, Male, Middle Aged, Mutation, Neoplasm Recurrence, Local genetics, Polymerase Chain Reaction, Temozolomide, Tumor Suppressor Proteins biosynthesis, Tumor Suppressor Proteins drug effects, Antineoplastic Agents, Alkylating adverse effects, Brain Neoplasms genetics, DNA-Binding Proteins drug effects, DNA-Binding Proteins genetics, Dacarbazine analogs & derivatives, Glioblastoma genetics
- Abstract
Purpose: Glioblastomas are treated by surgical resection followed by radiotherapy [X-ray therapy (XRT)] and the alkylating chemotherapeutic agent temozolomide. Recently, inactivating mutations in the mismatch repair gene MSH6 were identified in two glioblastomas recurrent post-temozolomide. Because mismatch repair pathway inactivation is a known mediator of alkylator resistance in vitro, these findings suggested that MSH6 inactivation was causally linked to these two recurrences. However, the extent of involvement of MSH6 in glioblastoma is unknown. We sought to determine the overall frequency and clinical relevance of MSH6 alterations in glioblastomas., Experimental Design: The MSH6 gene was sequenced in 54 glioblastomas. MSH6 and O(6)-methylguanine methyltransferase (MGMT) immunohistochemistry was systematically scored in a panel of 46 clinically well-characterized glioblastomas, and the corresponding patient response to treatment evaluated., Results: MSH6 mutation was not observed in any pretreatment glioblastoma (0 of 40), whereas 3 of 14 recurrent cases had somatic mutations (P = 0.015). MSH6 protein expression was detected in all pretreatment (17 of 17) cases examined but, notably, expression was lost in 7 of 17 (41%) recurrences from matched post-XRT + temozolomide cases (P = 0.016). Loss of MSH6 was not associated with O(6)-methylguanine methyltransferase status. Measurements of in vivo tumor growth using three-dimensional reconstructed magnetic resonance imaging showed that MSH6-negative glioblastomas had a markedly increased rate of growth while under temozolomide treatment (3.17 versus 0.04 cc/mo for MSH6-positive tumors; P = 0.020)., Conclusions: Loss of MSH6 occurs in a subset of post-XRT + temozolomide glioblastoma recurrences and is associated with tumor progression during temozolomide treatment, mirroring the alkylator resistance conferred by MSH6 inactivation in vitro. MSH6 deficiency may therefore contribute to the emergence of recurrent glioblastomas during temozolomide treatment.
- Published
- 2007
- Full Text
- View/download PDF
27. A hypermutation phenotype and somatic MSH6 mutations in recurrent human malignant gliomas after alkylator chemotherapy.
- Author
-
Hunter C, Smith R, Cahill DP, Stephens P, Stevens C, Teague J, Greenman C, Edkins S, Bignell G, Davies H, O'Meara S, Parker A, Avis T, Barthorpe S, Brackenbury L, Buck G, Butler A, Clements J, Cole J, Dicks E, Forbes S, Gorton M, Gray K, Halliday K, Harrison R, Hills K, Hinton J, Jenkinson A, Jones D, Kosmidou V, Laman R, Lugg R, Menzies A, Perry J, Petty R, Raine K, Richardson D, Shepherd R, Small A, Solomon H, Tofts C, Varian J, West S, Widaa S, Yates A, Easton DF, Riggins G, Roy JE, Levine KK, Mueller W, Batchelor TT, Louis DN, Stratton MR, Futreal PA, and Wooster R
- Subjects
- Aged, Brain Neoplasms drug therapy, Brain Neoplasms enzymology, Dacarbazine therapeutic use, Female, Glioma drug therapy, Glioma enzymology, Humans, Male, Middle Aged, Neoplasm Recurrence, Local enzymology, Protein Kinases genetics, Temozolomide, Antineoplastic Agents, Alkylating therapeutic use, Brain Neoplasms genetics, DNA-Binding Proteins genetics, Dacarbazine analogs & derivatives, Glioma genetics, Mutation, Neoplasm Recurrence, Local genetics
- Abstract
Malignant gliomas have a very poor prognosis. The current standard of care for these cancers consists of extended adjuvant treatment with the alkylating agent temozolomide after surgical resection and radiotherapy. Although a statistically significant increase in survival has been reported with this regimen, nearly all gliomas recur and become insensitive to further treatment with this class of agents. We sequenced 500 kb of genomic DNA corresponding to the kinase domains of 518 protein kinases in each of nine gliomas. Large numbers of somatic mutations were observed in two gliomas recurrent after alkylating agent treatment. The pattern of mutations in these cases showed strong similarity to that induced by alkylating agents in experimental systems. Further investigation revealed inactivating somatic mutations of the mismatch repair gene MSH6 in each case. We propose that inactivating somatic mutations of MSH6 confer resistance to alkylating agents in gliomas in vivo and concurrently unleash accelerated mutagenesis in resistant clones as a consequence of continued exposure to alkylating agents in the presence of defective mismatch repair. The evidence therefore suggests that when MSH6 is inactivated in gliomas, alkylating agents convert from induction of tumor cell death to promotion of neoplastic progression. These observations highlight the potential of large scale sequencing for revealing and elucidating mutagenic processes operative in individual human cancers.
- Published
- 2006
- Full Text
- View/download PDF
28. Three classes of genes mutated in colorectal cancers with chromosomal instability.
- Author
-
Wang Z, Cummins JM, Shen D, Cahill DP, Jallepalli PV, Wang TL, Parsons DW, Traverso G, Awad M, Silliman N, Ptak J, Szabo S, Willson JK, Markowitz SD, Goldberg ML, Karess R, Kinzler KW, Vogelstein B, Velculescu VE, and Lengauer C
- Subjects
- Animals, Cell Cycle Proteins genetics, DNA-Binding Proteins genetics, Drosophila melanogaster genetics, Female, Humans, MRE11 Homologue Protein, Polycomb Repressive Complex 1, Chromosomal Instability, Colorectal Neoplasms genetics, Mutation
- Abstract
Although most colorectal cancers are chromosomally unstable, the basis for this instability has not been defined. To determine whether genes shown to cause chromosomal instability in model systems were mutated in colorectal cancers, we identified their human homologues and determined their sequence in a panel of colorectal cancers. We found 19 somatic mutations in five genes representing three distinct instability pathways. Seven mutations were found in MRE11, whose product is involved in double-strand break repair. Four mutations were found among hZw10, hZwilch/FLJ10036, and hRod/KNTC, whose products bind to one another in a complex that localizes to kinetochores and controls chromosome segregation. Eight mutations were found in Ding, a previously uncharacterized gene with sequence similarity to the Saccharomyces cerevisiae Pds1, whose product is essential for proper chromosome disjunction. This analysis buttresses the evidence that chromosomal instability has a genetic basis and provides clues to the mechanistic basis of instability in cancers.
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.