19 results on '"Subapanditha M"'
Search Results
2. Hitting more birds with one stone: CD70 as an actionable immunotherapeutic target in recurrent glioblastoma
- Author
-
Seyfrid, M, primary, Maich, W, additional, Shaikh, MV, additional, Tatari, N, additional, Upreti, D, additional, Piyasena, D, additional, Subapanditha, M, additional, Savage, N, additional, McKenna, D, additional, Kuhlmann, L, additional, Khoo, A, additional, Salim, SK, additional, Bassey-Archibong, B, additional, Gwynne, W, additional, Chokshi, C, additional, Brown, K, additional, Murtaza, N, additional, Bakhshinyan, D, additional, Vora, P, additional, Venugopal, C, additional, Moffat, J, additional, and Singh, SK, additional
- Published
- 2021
- Full Text
- View/download PDF
3. PS2 - 172 Preclinical Validation of a Novel CD33/CD3 Bispecific T-Cell Engager (BiTE) Antibody to Target Patient-Derived Glioblastoma Cells
- Author
-
Vora, P., primary, Venugopal, C., additional, Choksi, C., additional, Qazi, M., additional, Adams, J., additional, London, M., additional, Subapanditha, M., additional, Singh, M., additional, Bakshinyan, D., additional, Sidhu, S., additional, Moffat, J., additional, and Singh, S., additional
- Published
- 2016
- Full Text
- View/download PDF
4. PS1 - 170 Bmi1 is a Therapeutic Target in Recurrent Childhood Medulloblastoma
- Author
-
Garg, N., primary, Bakhshinyan, D., additional, Manoranjan, B., additional, Venugopal, C., additional, Hallett, R., additional, Mahendram, S., additional, Vijayakumar, T., additional, Subapanditha, M., additional, Qazi, M., additional, Singh, M., additional, McFarlane, N., additional, Mann, A., additional, Vora, P., additional, Davis, T., additional, and Singh, S., additional
- Published
- 2016
- Full Text
- View/download PDF
5. PS2 - 171 Bmi1 Identifies Treatment-Refractory Stem Cells in Human Glioblastoma
- Author
-
Vora, P., primary, Qazi, M., additional, Venugopal, C., additional, Subapanditha, M., additional, Mahendram, S., additional, Chokshi, C., additional, Singh, M., additional, Bakhshinyan, D., additional, McFarlane, N., additional, and Singh, S., additional
- Published
- 2016
- Full Text
- View/download PDF
6. De novo GTP synthesis is a metabolic vulnerability for the interception of brain metastases.
- Author
-
Kieliszek AM, Mobilio D, Bassey-Archibong BI, Johnson JW, Piotrowski ML, de Araujo ED, Sedighi A, Aghaei N, Escudero L, Ang P, Gwynne WD, Zhang C, Quaile A, McKenna D, Subapanditha M, Tokar T, Vaseem Shaikh M, Zhai K, Chafe SC, Gunning PT, Montenegro-Burke JR, Venugopal C, Magolan J, and Singh SK
- Subjects
- Humans, Animals, Cell Line, Tumor, Mice, Cell Proliferation, Female, Brain Neoplasms secondary, Brain Neoplasms metabolism, Brain Neoplasms genetics, Brain Neoplasms pathology, IMP Dehydrogenase metabolism, IMP Dehydrogenase genetics, Guanosine Triphosphate metabolism
- Abstract
Patients with brain metastases (BM) face a 90% mortality rate within one year of diagnosis and the current standard of care is palliative. Targeting BM-initiating cells (BMICs) is a feasible strategy to treat BM, but druggable targets are limited. Here, we apply Connectivity Map analysis to lung-, breast-, and melanoma-pre-metastatic BMIC gene expression signatures and identify inosine monophosphate dehydrogenase (IMPDH), the rate-limiting enzyme in the de novo GTP synthesis pathway, as a target for BM. We show that pharmacological and genetic perturbation of IMPDH attenuates BMIC proliferation in vitro and the formation of BM in vivo. Metabolomic analyses and CRISPR knockout studies confirm that de novo GTP synthesis is a potent metabolic vulnerability in BM. Overall, our work employs a phenotype-guided therapeutic strategy to uncover IMPDH as a relevant target for attenuating BM outgrowth, which may provide an alternative treatment strategy for patients who are otherwise limited to palliation., Competing Interests: Declaration of interests A.M.K., J.W.J., C.V., J.M., and S.K.S. are listed as co-inventors for a PCT patent that has been filed, relating to this work., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
7. Targeting axonal guidance dependencies in glioblastoma with ROBO1 CAR T cells.
- Author
-
Chokshi CR, Shaikh MV, Brakel B, Rossotti MA, Tieu D, Maich W, Anand A, Chafe SC, Zhai K, Suk Y, Kieliszek AM, Miletic P, Mikolajewicz N, Chen D, McNicol JD, Chan K, Tong AHY, Kuhlmann L, Liu L, Alizada Z, Mobilio D, Tatari N, Savage N, Aghaei N, Grewal S, Puri A, Subapanditha M, McKenna D, Ignatchenko V, Salamoun JM, Kwiecien JM, Wipf P, Sharlow ER, Provias JP, Lu JQ, Lazo JS, Kislinger T, Lu Y, Brown KR, Venugopal C, Henry KA, Moffat J, and Singh SK
- Subjects
- Animals, Female, Humans, Mice, Cell Line, Tumor, Immunotherapy, Adoptive methods, Neoplasm Recurrence, Local pathology, Neoplasm Recurrence, Local genetics, Nerve Tissue Proteins metabolism, Nerve Tissue Proteins genetics, Protein Tyrosine Phosphatases genetics, Protein Tyrosine Phosphatases metabolism, Receptors, Chimeric Antigen immunology, Receptors, Chimeric Antigen genetics, Receptors, Chimeric Antigen metabolism, Signal Transduction, T-Lymphocytes immunology, Xenograft Model Antitumor Assays, Brain Neoplasms pathology, Brain Neoplasms immunology, Brain Neoplasms genetics, Glioblastoma pathology, Glioblastoma genetics, Glioblastoma immunology, Receptors, Immunologic metabolism, Receptors, Immunologic genetics, Roundabout Proteins antagonists & inhibitors
- Abstract
Resistance to genotoxic therapies and tumor recurrence are hallmarks of glioblastoma (GBM), an aggressive brain tumor. In this study, we investigated functional drivers of post-treatment recurrent GBM through integrative genomic analyses, genome-wide genetic perturbation screens in patient-derived GBM models and independent lines of validation. Specific genetic dependencies were found consistent across recurrent tumor models, accompanied by increased mutational burden and differential transcript and protein expression compared to its primary GBM predecessor. Our observations suggest a multi-layered genetic response to drive tumor recurrence and implicate PTP4A2 (protein tyrosine phosphatase 4A2) as a modulator of self-renewal, proliferation and tumorigenicity in recurrent GBM. Genetic perturbation or small-molecule inhibition of PTP4A2 acts through a dephosphorylation axis with roundabout guidance receptor 1 (ROBO1) and its downstream molecular players, exploiting a functional dependency on ROBO signaling. Because a pan-PTP4A inhibitor was limited by poor penetrance across the blood-brain barrier in vivo, we engineered a second-generation chimeric antigen receptor (CAR) T cell therapy against ROBO1, a cell surface receptor enriched across recurrent GBM specimens. A single dose of ROBO1-targeted CAR T cells doubled median survival in cell-line-derived xenograft (CDX) models of recurrent GBM. Moreover, in CDX models of adult lung-to-brain metastases and pediatric relapsed medulloblastoma, ROBO1 CAR T cells eradicated tumors in 50-100% of mice. Our study identifies a promising multi-targetable PTP4A-ROBO1 signaling axis that drives tumorigenicity in recurrent GBM, with potential in other malignant brain tumors., (© 2024. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2024
- Full Text
- View/download PDF
8. An HLA-G/SPAG9/STAT3 axis promotes brain metastases.
- Author
-
Bassey-Archibong BI, Rajendra Chokshi C, Aghaei N, Kieliszek AM, Tatari N, McKenna D, Singh M, Kalpana Subapanditha M, Parmar A, Mobilio D, Savage N, Lam F, Tokar T, Provias J, Lu Y, Chafe SC, Swanton C, Hynds RE, Venugopal C, and Singh SK
- Subjects
- Adult, Humans, Adaptor Proteins, Signal Transducing, Brain pathology, Lung pathology, STAT3 Transcription Factor genetics, Brain Neoplasms secondary, HLA-G Antigens genetics, Lung Neoplasms pathology, Melanoma pathology, Breast Neoplasms pathology
- Abstract
Brain metastases (BM) are the most common brain neoplasm in adults. Current BM therapies still offer limited efficacy and reduced survival outcomes, emphasizing the need for a better understanding of the disease. Herein, we analyzed the transcriptional profile of brain metastasis initiating cells (BMICs) at two distinct stages of the brain metastatic cascade-the "premetastatic" or early stage when they first colonize the brain and the established macrometastatic stage. RNA sequencing was used to obtain the transcriptional profiles of premetastatic and macrometastatic (non-premetastatic) lung, breast, and melanoma BMICs. We identified that lung, breast, and melanoma premetastatic BMICs share a common transcriptomic signature that is distinct from their non-premetastatic counterparts. Importantly, we show that premetastatic BMICs exhibit increased expression of HLA-G, which we further demonstrate functions in an HLA-G/SPAG9/STAT3 axis to promote the establishment of brain metastatic lesions. Our findings suggest that unraveling the molecular landscape of premetastatic BMICs allows for the identification of clinically relevant targets that can possibly inform the development of preventive and/or more efficacious BM therapies.
- Published
- 2023
- Full Text
- View/download PDF
9. Author Correction: Characterization of an RNA binding protein interactome reveals a context-specific post-transcriptional landscape of MYC-amplified medulloblastoma.
- Author
-
Kameda-Smith MM, Zhu H, Luo EC, Suk Y, Xella A, Yee B, Chokshi C, Xing S, Tan F, Fox RG, Adile AA, Bakhshinyan D, Brown K, Gwynne WD, Subapanditha M, Miletic P, Picard D, Burns I, Moffat J, Paruch K, Fleming A, Hope K, Provias JP, Remke M, Lu Y, Reya T, Venugopal C, Reimand J, Wechsler-Reya RJ, Yeo GW, and Singh SK
- Published
- 2023
- Full Text
- View/download PDF
10. Characterization of an RNA binding protein interactome reveals a context-specific post-transcriptional landscape of MYC-amplified medulloblastoma.
- Author
-
Kameda-Smith MM, Zhu H, Luo EC, Suk Y, Xella A, Yee B, Chokshi C, Xing S, Tan F, Fox RG, Adile AA, Bakhshinyan D, Brown K, Gwynne WD, Subapanditha M, Miletic P, Picard D, Burns I, Moffat J, Paruch K, Fleming A, Hope K, Provias JP, Remke M, Lu Y, Reya T, Venugopal C, Reimand J, Wechsler-Reya RJ, Yeo GW, and Singh SK
- Subjects
- Animals, Mice, Humans, Proteomics, RNA-Binding Proteins genetics, Nerve Tissue Proteins, Medulloblastoma genetics, Brain Neoplasms, Cerebellar Neoplasms genetics
- Abstract
Pediatric medulloblastoma (MB) is the most common solid malignant brain neoplasm, with Group 3 (G3) MB representing the most aggressive subgroup. MYC amplification is an independent poor prognostic factor in G3 MB, however, therapeutic targeting of the MYC pathway remains limited and alternative therapies for G3 MB are urgently needed. Here we show that the RNA-binding protein, Musashi-1 (MSI1) is an essential mediator of G3 MB in both MYC-overexpressing mouse models and patient-derived xenografts. MSI1 inhibition abrogates tumor initiation and significantly prolongs survival in both models. We identify binding targets of MSI1 in normal neural and G3 MB stem cells and then cross referenced these data with unbiased large-scale screens at the transcriptomic, translatomic and proteomic levels to systematically dissect its functional role. Comparative integrative multi-omic analyses of these large datasets reveal cancer-selective MSI1-bound targets sharing multiple MYC associated pathways, providing a valuable resource for context-specific therapeutic targeting of G3 MB., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
11. Dual Antigen T Cell Engagers Targeting CA9 as an Effective Immunotherapeutic Modality for Targeting CA9 in Solid Tumors.
- Author
-
Tatari N, Zhang X, Chafe SC, McKenna D, Lawson KA, Subapanditha M, Shaikh MV, Seyfrid M, Savage N, Venugopal C, Moffat J, and Singh SK
- Subjects
- Adult, Antigens, Neoplasm therapeutic use, Carbonic Anhydrase IX metabolism, Humans, Hypoxia, Immunotherapy, T-Lymphocytes metabolism, Brain Neoplasms metabolism, Carbonic Anhydrases metabolism, Carbonic Anhydrases therapeutic use, Carcinoma, Renal Cell therapy, Glioblastoma therapy, Kidney Neoplasms therapy
- Abstract
Glioblastomas (GBM), the most common malignant primary adult brain tumors, are uniformly lethal and are in need of improved therapeutic modalities. GBM contain extensive regions of hypoxia and are enriched in therapy resistant brain tumor-initiating cells (BTICs). Carbonic anhydrase 9 (CA9) is a hypoxia-induced cell surface enzyme that plays an important role in maintenance of stem cell survival and therapeutic resistance. Here we demonstrate that CA9 is highly expressed in patient-derived BTICs. CA9
+ GBM BTICs showed increased self-renewal and proliferative capacity. To target CA9, we developed dual antigen T cell engagers (DATEs) that were exquisitely specific for CA9-positive patient-derived clear cell Renal Cell Carcinoma (ccRCC) and GBM cells. Combined treatment of either ccRCC or GBM cells with the CA9 DATE and T cells resulted in T cell activation, increased release of pro-inflammatory cytokines and enhanced cytotoxicity in a CA9-dependent manner. Treatment of ccRCC and GBM patient-derived xenografts markedly reduced tumor burden and extended survival. These data suggest that the CA9 DATE could provide a novel therapeutic strategy for patients with solid tumors expressing CA9 to overcome treatment resistance. ., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Tatari, Zhang, Chafe, McKenna, Lawson, Subapanditha, Shaikh, Seyfrid, Savage, Venugopal, Moffat and Singh.)- Published
- 2022
- Full Text
- View/download PDF
12. CD70 as an actionable immunotherapeutic target in recurrent glioblastoma and its microenvironment.
- Author
-
Seyfrid M, Maich WT, Shaikh VM, Tatari N, Upreti D, Piyasena D, Subapanditha M, Savage N, McKenna D, Mikolajewicz N, Han H, Chokshi C, Kuhlmann L, Khoo A, Salim SK, Archibong-Bassey B, Gwynne W, Brown K, Murtaza N, Bakhshinyan D, Vora P, Venugopal C, Moffat J, Kislinger T, and Singh S
- Subjects
- Animals, Brain Neoplasms immunology, Cell Proliferation, Glioblastoma immunology, Humans, Male, Mice, Inbred NOD, Mice, SCID, Neoplasm Recurrence, Local, Prognosis, Mice, Brain Neoplasms therapy, CD27 Ligand metabolism, Glioblastoma therapy, Immunotherapy methods, Proteomics methods, Transcriptome genetics, Tumor Microenvironment immunology
- Abstract
Purpose: Glioblastoma (GBM) patients suffer from a dismal prognosis, with standard of care therapy inevitably leading to therapy-resistant recurrent tumors. The presence of cancer stem cells (CSCs) drives the extensive heterogeneity seen in GBM, prompting the need for novel therapies specifically targeting this subset of tumor-driving cells. Here, we identify CD70 as a potential therapeutic target for recurrent GBM CSCs., Experimental Design: In the current study, we identified the relevance and functional influence of CD70 on primary and recurrent GBM cells, and further define its function using established stem cell assays. We use CD70 knockdown studies, subsequent RNAseq pathway analysis, and in vivo xenotransplantation to validate CD70's role in GBM. Next, we developed and tested an anti-CD70 chimeric antigen receptor (CAR)-T therapy, which we validated in vitro and in vivo using our established preclinical model of human GBM. Lastly, we explored the importance of CD70 in the tumor immune microenvironment (TIME) by assessing the presence of its receptor, CD27, in immune infiltrates derived from freshly resected GBM tumor samples., Results: CD70 expression is elevated in recurrent GBM and CD70 knockdown reduces tumorigenicity in vitro and in vivo . CD70 CAR-T therapy significantly improves prognosis in vivo . We also found CD27 to be present on the cell surface of multiple relevant GBM TIME cell populations, notably putative M1 macrophages and CD4 T cells., Conclusion: CD70 plays a key role in recurrent GBM cell aggressiveness and maintenance. Immunotherapeutic targeting of CD70 significantly improves survival in animal models and the CD70/CD27 axis may be a viable polytherapeutic avenue to co-target both GBM and its TIME., Competing Interests: Competing interests: DU, DB and PV are employees of Century Therapeutics Canada. The other authors declare no competing interests., (© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2022
- Full Text
- View/download PDF
13. Wnt activation as a therapeutic strategy in medulloblastoma.
- Author
-
Manoranjan B, Venugopal C, Bakhshinyan D, Adile AA, Richards L, Kameda-Smith MM, Whitley O, Dvorkin-Gheva A, Subapanditha M, Savage N, Tatari N, McKenna D, Bassey-Archibong B, Winegarden N, Hallett R, Provias JP, Yarascavitch B, Ajani O, Fleming A, Bader GD, Pugh TJ, Doble BW, and Singh SK
- Subjects
- Animals, Carcinogenesis, Cell Line, Tumor, Cell Proliferation, Cerebellar Neoplasms pathology, Disease Models, Animal, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Heterografts, Humans, Medulloblastoma genetics, Medulloblastoma pathology, Mice, Stem Cells, Wnt Proteins genetics, Wnt Signaling Pathway, beta Catenin therapeutic use, Cerebellar Neoplasms therapy, Medulloblastoma therapy, Wnt Proteins pharmacology, Wnt Proteins therapeutic use
- Abstract
Medulloblastoma (MB) is defined by four molecular subgroups (Wnt, Shh, Group 3, Group 4) with Wnt MB having the most favorable prognosis. Since prior reports have illustrated the antitumorigenic role of Wnt activation in Shh MB, we aimed to assess the effects of activated canonical Wnt signaling in Group 3 and 4 MBs. By using primary patient-derived MB brain tumor-initiating cell (BTIC) lines, we characterize differences in the tumor-initiating capacity of Wnt, Group 3, and Group 4 MB. With single cell RNA-seq technology, we demonstrate the presence of rare Wnt-active cells in non-Wnt MBs, which functionally retain the impaired tumorigenic potential of Wnt MB. In treating MB xenografts with a Wnt agonist, we provide a rational therapeutic option in which the protective effects of Wnt-driven MBs may be augmented in Group 3 and 4 MB and thereby support emerging data for a context-dependent tumor suppressive role for Wnt/β-catenin signaling.
- Published
- 2020
- Full Text
- View/download PDF
14. The Rational Development of CD133-Targeting Immunotherapies for Glioblastoma.
- Author
-
Vora P, Venugopal C, Salim SK, Tatari N, Bakhshinyan D, Singh M, Seyfrid M, Upreti D, Rentas S, Wong N, Williams R, Qazi MA, Chokshi C, Ding A, Subapanditha M, Savage N, Mahendram S, Ford E, Adile AA, McKenna D, McFarlane N, Huynh V, Wylie RG, Pan J, Bramson J, Hope K, Moffat J, and Singh S
- Subjects
- AC133 Antigen, Animals, Humans, Immunotherapy, Mice, Neoplastic Stem Cells, Brain Neoplasms therapy, Glioblastoma therapy
- Abstract
CD133 marks self-renewing cancer stem cells (CSCs) in a variety of solid tumors, and CD133+ tumor-initiating cells are known markers of chemo- and radio-resistance in multiple aggressive cancers, including glioblastoma (GBM), that may drive intra-tumoral heterogeneity. Here, we report three immunotherapeutic modalities based on a human anti-CD133 antibody fragment that targets a unique epitope present in glycosylated and non-glycosylated CD133 and studied their effects on targeting CD133+ cells in patient-derived models of GBM. We generated an immunoglobulin G (IgG) (RW03-IgG), a dual-antigen T cell engager (DATE), and a CD133-specific chimeric antigen receptor T cell (CAR-T): CART133. All three showed activity against patient-derived CD133+ GBM cells, and CART133 cells demonstrated superior efficacy in patient-derived GBM xenograft models without causing adverse effects on normal CD133+ hematopoietic stem cells in humanized CD34+ mice. Thus, CART133 cells may be a therapeutically tractable strategy to target CD133+ CSCs in human GBM or other treatment-resistant primary cancers., Competing Interests: Declaration of Interests P.V., C.V., R.W., J.M., and S.S. have patents around CD133 binding agents and uses thereof. These patents include Canadian Patent Application no. 2,962,157; Chinese Patent Application no. 2017800782373; European Patent Application no. EP17863201; Japanese Patent Application no. 2019-521046; and U.S. Patent Application no. 16/342,807. P.V., C.V., J.M., and S.S. are also shareholders of Empirica Therapeutics, which has an exclusive license to the IP as mentioned above. P.V. and D.B. are employees of Empirica Therapeutics., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
15. A CD133-AKT-Wnt signaling axis drives glioblastoma brain tumor-initiating cells.
- Author
-
Manoranjan B, Chokshi C, Venugopal C, Subapanditha M, Savage N, Tatari N, Provias JP, Murty NK, Moffat J, Doble BW, and Singh SK
- Subjects
- Cell Line, Tumor, Glioblastoma metabolism, Humans, AC133 Antigen metabolism, Carcinogenesis, Glioblastoma pathology, Proto-Oncogene Proteins c-akt metabolism, Wnt Signaling Pathway
- Abstract
Mechanistic insight into signaling pathways downstream of surface receptors has been revolutionized with integrated cancer genomics. This has fostered current treatment modalities, namely immunotherapy, to capitalize on targeting key oncogenic signaling nodes downstream of a limited number of surface markers. Unfortunately, rudimentary mechanistic understanding of most other cell surface proteins has reduced the clinical utility of these markers. CD133 has reproducibly been shown to correlate with disease progression, recurrence, and poor overall survivorship in the malignant adult brain tumor, glioblastoma (GBM). Using several patient-derived CD133
high and CD133low GBMs we describe intrinsic differences in determinants of stemness, which we owe to a CD133-AKT-Wnt signaling axis in which CD133 functions as a putative cell surface receptor for AKT-dependent Wnt activation. These findings may have implications for personalized oncology trials targeting PI3K/AKT or Wnt as both pathways may be activated independent of their canonical drivers, leading to treatment resistance and disease relapse.- Published
- 2020
- Full Text
- View/download PDF
16. Bmi1 regulates human glioblastoma stem cells through activation of differential gene networks in CD133+ brain tumor initiating cells.
- Author
-
Vora P, Seyfrid M, Venugopal C, Qazi MA, Salim S, Isserlin R, Subapanditha M, O'Farrell E, Mahendram S, Singh M, Bakhshinyan D, Chokshi C, McFarlane N, Dvorkin-Gheva A, Brown KR, Murty N, Moffat J, Bader GD, and Singh SK
- Subjects
- AC133 Antigen genetics, Animals, Apoptosis, Brain Neoplasms genetics, Brain Neoplasms metabolism, Cell Proliferation, Glioblastoma genetics, Glioblastoma metabolism, Humans, Mice, Mice, Inbred NOD, Mice, SCID, Neoplastic Stem Cells metabolism, Polycomb Repressive Complex 1 genetics, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, AC133 Antigen metabolism, Brain Neoplasms pathology, Gene Expression Regulation, Neoplastic, Gene Regulatory Networks, Glioblastoma pathology, Neoplastic Stem Cells pathology, Polycomb Repressive Complex 1 metabolism
- Abstract
Purpose: Glioblastoma (GBM) is the most aggressive adult brain cancer, with a 15 month median survivorship attributed to the existence of treatment-refractory brain tumor initiating cells (BTICs). In order to better understand the mechanisms regulating the tumorigenic properties of this population, we studied the role of the polycomb group member BMI1 in our patient-derived GBM BTICs and its relationship with CD133, a well-established marker of BTICs., Methods: Using gain and loss-of-function studies for Bmi1 in neural stem cells (NSCs) and patient-derived GBM BTICs respectively, we assessed in vitro self-renewal and in vivo tumor formation in these two cell populations. We further explored the BMI1 transcriptional regulatory network through RNA sequencing of different GBM BTIC populations that were knocked down for Bmi1., Results: There is a differential role of BMI1 in CD133-positive cells, notably involving cell metabolism. In addition, we identified pivotal targets downstream of BMI1 in CD133+ cells such as integrin alpha 2 (ITGA2), that may contribute to regulating GBM stem cell properties., Conclusions: Our work sheds light on the association of three genes with CD133-BMI1 circuitry, their importance as downstream effectors of the BMI1 signalling pathway, and their potential as future targets for tackling GBM treatment-resistant cell populations.
- Published
- 2019
- Full Text
- View/download PDF
17. TAp73 Modifies Metabolism and Positively Regulates Growth of Cancer Stem-Like Cells in a Redox-Sensitive Manner.
- Author
-
Sharif T, Dai C, Martell E, Ghassemi-Rad MS, Hanes MR, Murphy PJ, Kennedy BE, Venugopal C, Subapanditha M, Giacomantonio CA, Marcato P, Singh SK, and Gujar S
- Subjects
- Animals, Carcinogenesis genetics, Carcinogenesis metabolism, Cell Self Renewal genetics, Female, Gene Knockdown Techniques, Humans, Mice, Neoplasms pathology, Neoplastic Stem Cells pathology, Oxidation-Reduction, RNA, Small Interfering metabolism, Tumor Protein p73 genetics, Xenograft Model Antitumor Assays, Gene Expression Regulation, Neoplastic, Neoplasms genetics, Neoplastic Stem Cells metabolism, Tumor Protein p73 metabolism
- Abstract
Purpose: Stem-like cancer cells, with characteristic self-renewal abilities, remain highly refractory to various clinical interventions. As such, stemness-inhibiting entities, such as tumor suppressor p53, are therapeutically pursued for their anticancer activities. Interestingly, similar implications for tumor suppressor TAp73 in regulating stemness features within stem-like cancer cells remain unknown. Experimental Design: This study utilizes various in vitro molecular biology techniques, including immunoblotting, qRT-PCR, and mass spectrometry-based proteomics, and metabolomics approaches to study the role of TAp73 in human and murine embryonal carcinoma stem-like cells (ECSLC) as well as human breast cancer stem-like cells (BCSLC). These findings were confirmed using patient-derived brain tumor-initiating cells (BTIC) and in vivo xenograft models., Results: TAp73 inhibition decreases the expression of stem cell transcription factors Oct4, Nanog, and Sox-2, as well as tumorsphere formation capacity in ECSLCs. In vivo , TAp73-deficient ECSLCs and BCSLCs demonstrate decreased tumorigenic potential when xenografted in mice. Mechanistically, TAp73 modifies the proline regulatory axis through regulation of enzymes GLS, OAT , and PYCR1 involved in the interconversion of proline-glutamine-ornithine. Further, TAp73 deficiency exacerbates glutamine dependency, enhances accumulation of reactive oxygen species through reduced superoxide dismutase 1 (SOD1) expression, and promotes differentiation by arresting cell cycle and elevating autophagy. Most importantly, the knockdown of TAp73 in CD133
HI BTICs, separated from three different glioblastoma patients, strongly decreases the expression of prosurvival factors Sox-2, BMI-1, and SOD1, and profoundly decreases their self-renewal capacity as evidenced through their reduced tumorsphere formation ability., Conclusions: Collectively, we reveal a clinically relevant aspect of cancer cell growth and stemness regulation through TAp73-mediated redox-sensitive metabolic reprogramming., (©2018 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
18. BMI1 is a therapeutic target in recurrent medulloblastoma.
- Author
-
Bakhshinyan D, Venugopal C, Adile AA, Garg N, Manoranjan B, Hallett R, Wang X, Mahendram S, Vora P, Vijayakumar T, Subapanditha M, Singh M, Kameda-Smith MM, Qazi M, McFarlane N, Mann A, Ajani OA, Yarascavitch B, Ramaswamy V, Farooq H, Morrissy S, Cao L, Sydorenko N, Baiazitov R, Du W, Sheedy J, Weetall M, Moon YC, Lee CS, Kwiecien JM, Delaney KH, Doble B, Cho YJ, Mitra S, Kaplan D, Taylor MD, Davis TW, and Singh SK
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Cerebellar Neoplasms genetics, Cerebellar Neoplasms metabolism, Child, Epigenesis, Genetic, Gene Expression Regulation, Neoplastic drug effects, Humans, Medulloblastoma genetics, Medulloblastoma metabolism, Mice, Neoplastic Stem Cells cytology, Neoplastic Stem Cells metabolism, Polycomb Repressive Complex 1 genetics, Small Molecule Libraries pharmacology, Treatment Outcome, Up-Regulation drug effects, Xenograft Model Antitumor Assays, Cerebellar Neoplasms drug therapy, Medulloblastoma drug therapy, Neoplastic Stem Cells drug effects, Polycomb Repressive Complex 1 antagonists & inhibitors, Small Molecule Libraries administration & dosage
- Abstract
Medulloblastoma (MB) is the most frequent malignant pediatric brain tumor, representing 20% of newly diagnosed childhood central nervous system malignancies. Although advances in multimodal therapy yielded a 5-year survivorship of 80%, MB still accounts for the leading cause of childhood cancer mortality. In this work, we describe the epigenetic regulator BMI1 as a novel therapeutic target for the treatment of recurrent human Group 3 MB, a childhood brain tumor for which there is virtually no treatment option beyond palliation. Current clinical trials for recurrent MB patients based on genomic profiles of primary, treatment-naive tumors will provide limited clinical benefit since recurrent metastatic MBs are highly genetically divergent from their primary tumor. Using a small molecule inhibitor against BMI1, PTC-028, we were able to demonstrate complete ablation of self-renewal of MB stem cells in vitro. When administered to mice xenografted with patient tumors, we observed significant reduction in tumor burden in both local and metastatic compartments and subsequent increased survival, without neurotoxicity. Strikingly, serial in vivo re-transplantation assays demonstrated a marked reduction in tumor initiation ability of recurrent MB cells upon re-transplantation of PTC-028-treated cells into secondary recipient mouse brains. As Group 3 MB is often metastatic and uniformly fatal at recurrence, with no current or planned trials of targeted therapy, an efficacious targeted agent would be rapidly transitioned to clinical trials.
- Published
- 2019
- Full Text
- View/download PDF
19. Pyrvinium Targets CD133 in Human Glioblastoma Brain Tumor-Initiating Cells.
- Author
-
Venugopal C, Hallett R, Vora P, Manoranjan B, Mahendram S, Qazi MA, McFarlane N, Subapanditha M, Nolte SM, Singh M, Bakhshinyan D, Garg N, Vijayakumar T, Lach B, Provias JP, Reddy K, Murty NK, Doble BW, Bhatia M, Hassell JA, and Singh SK
- Subjects
- AC133 Antigen, Animals, Antigens, CD genetics, Antigens, CD metabolism, Brain Neoplasms drug therapy, Brain Neoplasms genetics, Brain Neoplasms mortality, Cell Proliferation, Cell Self Renewal drug effects, Cell Self Renewal genetics, Disease Models, Animal, Gene Expression, Gene Expression Profiling, Gene Expression Regulation, Neoplastic drug effects, Gene Knockdown Techniques, Gene Regulatory Networks, Glioblastoma drug therapy, Glioblastoma genetics, Glioblastoma mortality, Glycoproteins genetics, Glycoproteins metabolism, Humans, Peptides genetics, Peptides metabolism, Prognosis, Signal Transduction drug effects, Spheroids, Cellular, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Brain Neoplasms metabolism, Glioblastoma metabolism, Glycoproteins antagonists & inhibitors, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells metabolism, Peptides antagonists & inhibitors, Pyrvinium Compounds pharmacology
- Abstract
Purpose: Clonal evolution of cancer may be regulated by determinants of stemness, specifically self-renewal, and current therapies have not considered how genetic perturbations or properties of stemness affect such functional processes. Glioblastoma-initiating cells (GICs), identified by expression of the cell surface marker CD133, are shown to be chemoradioresistant. In the current study, we sought to elucidate the functional role of CD133 in self-renewal and identify compounds that can specifically target this CD133(+) treatment-refractory population., Experimental Design: Using gain/loss-of-function studies for CD133 we assessed the in vitro self-renewal and in vivo tumor formation capabilities of patient-derived glioblastoma cells. We generated a CD133 signature combined with an in silico screen to find compounds that target GICs. Self-renewal and proliferation assays on CD133-sorted samples were performed to identify the preferential action of hit compounds. In vivo efficacy of the lead compound pyrvinium was assessed in intracranial GIC xenografts and survival studies. Lastly, microarray analysis was performed on pyrvinium-treated GICs to discover core signaling events involved., Results: We discovered pyrvinium, a small-molecule inhibitor of GIC self-renewal in vitro and in vivo, in part through inhibition of Wnt/β-catenin signaling and other essential stem cell regulatory pathways. We provide a therapeutically tractable strategy to target self-renewing, chemoradioresistant, and functionally important CD133(+) stem cells that drive glioblastoma relapse and mortality., Conclusions: Our study provides an integrated approach for the eradication of clonal populations responsible for cancer progression, and may apply to other aggressive and heterogeneous cancers., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.