209 results on '"Emdad L"'
Search Results
2. Detecting Tumor Metastases
- Author
-
Menezes, M.E., primary, Das, S.K., additional, Minn, I., additional, Emdad, L., additional, Wang, X.-Y., additional, Sarkar, D., additional, Pomper, M.G., additional, and Fisher, P.B., additional
- Published
- 2016
- Full Text
- View/download PDF
3. Evolving Strategies for Therapeutically Targeting Cancer Stem Cells
- Author
-
Talukdar, S., primary, Emdad, L., additional, Das, S.K., additional, Sarkar, D., additional, and Fisher, P.B., additional
- Published
- 2016
- Full Text
- View/download PDF
4. Embryonic stem cell (ESC)-mediated transgene delivery induces growth suppression, apoptosis and radiosensitization, and overcomes temozolomide resistance in malignant gliomas
- Author
-
Germano, I M, Emdad, L, Qadeer, Z A, Binello, E, and Uzzaman, M
- Published
- 2010
- Full Text
- View/download PDF
5. Astrocyte elevated gene-1 activates cell survival pathways through PI3K-Akt signaling
- Author
-
Lee, S-G, Su, Z-Z, Emdad, L, Sarkar, D, Franke, T F, and Fisher, P B
- Published
- 2008
- Full Text
- View/download PDF
6. Targeting inhibition of K-ras enhances Ad.mda-7-induced growth suppression and apoptosis in mutant K-ras colorectal cancer cells
- Author
-
Lebedeva, I V, Su, Z-Z, Emdad, L, Kolomeyer, A, Sarkar, D, Kitada, S, Waxman, S, Reed, J C, and Fisher, P B
- Published
- 2007
- Full Text
- View/download PDF
7. Ionizing radiation enhances therapeutic activity of mda-7/IL-24: overcoming radiation- and mda-7/IL-24-resistance in prostate cancer cells overexpressing the antiapoptotic proteins bcl-xL or bcl-2
- Author
-
Su, Z-Z, Lebedeva, I V, Sarkar, D, Emdad, L, Gupta, P, Kitada, S, Dent, P, Reed, J C, and Fisher, P B
- Published
- 2006
- Full Text
- View/download PDF
8. Detecting Tumor Metastases: The Road to Therapy Starts Here
- Author
-
Menezes, M.E., Das, S.K., Minn, I., Emdad, L., Wang, X.-Y., Sarkar, D., Pomper, M.G., and Fisher, P.B.
- Subjects
Neoplasms ,Disease Progression ,Animals ,Humans ,Antineoplastic Agents ,Neoplasm Metastasis ,Article - Abstract
Metastasis is the complex process by which primary tumor cells migrate and establish secondary tumors in an adjacent or distant location in the body. Early detection of metastatic disease and effective therapeutic options for targeting these detected metastases remain impediments to effectively treating patients with advanced cancers. If metastatic lesions are identified early, patients might maximally benefit from effective early therapeutic interventions. Further, monitoring patients whose primary tumors are effectively treated for potential metastatic disease onset is also highly valuable. Finally, patients with metastatic disease can be monitored for efficacy of specific therapeutic interventions through effective metastatic detection techniques. Thus, being able to detect and visualize metastatic lesions is key and provides potential to greatly improve overall patient outcomes. In order to achieve these objectives, researchers have endeavored to mechanistically define the steps involved in the metastatic process as well as ways to effectively detect metastatic progression. We presently overview various preclinical and clinical in vitro and in vivo assays developed to more efficiently detect tumor metastases, which provides the foundation for developing more effective therapies for this invariably fatal component of the cancerous process.
- Published
- 2016
9. Enterovirus 2Apro targets MDA5 and MAVS in infected cells
- Author
-
Feng, Q., Langereis, M.A., Lork, M., Nguyen, M.H., Hato, S.V., Lanke, K.H.W., Emdad, L., Bhoopathi, P., Fisher, P.B., Lloyd, R.E., Kuppeveld, F.J.M. van, Feng, Q., Langereis, M.A., Lork, M., Nguyen, M.H., Hato, S.V., Lanke, K.H.W., Emdad, L., Bhoopathi, P., Fisher, P.B., Lloyd, R.E., and Kuppeveld, F.J.M. van
- Abstract
Contains fulltext : 137931.pdf (publisher's version ) (Open Access), RIG-I-like receptors (RLRs) MDA5 and RIG-I are key players in the innate antiviral response. Upon recognition of viral RNA, they interact with MAVS, eventually inducing type I interferon production. The interferon induction pathway is commonly targeted by viruses. How enteroviruses suppress interferon production is incompletely understood. MDA5 has been suggested to undergo caspase- and proteasome-mediated degradation during poliovirus infection. Additionally, MAVS is reported to be cleaved during infection with coxsackievirus B3 (CVB3) by the CVB3 proteinase 3C(pro), whereas MAVS cleavage by enterovirus 71 has been attributed to 2A(pro). As yet, a detailed examination of the RLR pathway as a whole during any enterovirus infection is lacking. We performed a comprehensive analysis of crucial factors of the RLR pathway, including MDA5, RIG-I, LGP2, MAVS, TBK1, and IRF3, during infection of CVB3, a human enterovirus B (HEV-B) species member. We show that CVB3 inhibits the RLR pathway upstream of TBK1 activation, as demonstrated by limited phosphorylation of TBK1 and a lack of IRF3 phosphorylation. Furthermore, we show that MDA5, MAVS, and RIG-I all undergo proteolytic degradation in CVB3-infected cells through a caspase- and proteasome-independent manner. We convincingly show that MDA5 and MAVS cleavages are both mediated by CVB3 2A(pro), while RIG-I is cleaved by 3C(pro). Moreover, we show that proteinases 2A(pro) and 3C(pro) of poliovirus (HEV-C) and enterovirus 71 (HEV-A) exert the same functions. This study identifies a critical role of 2A(pro) by cleaving MDA5 and MAVS and shows that enteroviruses use a common strategy to counteract the interferon response in infected cells. IMPORTANCE: Human enteroviruses (HEVs) are important pathogens that cause a variety of diseases in humans, including poliomyelitis, hand, foot, and mouth disease, viral meningitis, cardiomyopathy, and more. Like many other viruses, enteroviruses target the host immune pathways to gain replicat
- Published
- 2014
10. Chemoprevention Gene Therapy (CGT) of Pancreatic Cancer Using Perillyl Alcohol and a Novel Chimeric Serotype Cancer Terminator Virus
- Author
-
Sarkar, S., primary, Azab, B., additional, Quinn, B.A., additional, Shen, X., additional, Dent, P., additional, Klibanov, A.L., additional, Emdad, L., additional, Das, S.K., additional, Sarkar, D., additional, and Fisher, P.B., additional
- Published
- 2014
- Full Text
- View/download PDF
11. Chemoprevention Gene Therapy (CGT): Novel Combinatorial Approach for Preventing and Treating Pancreatic Cancer
- Author
-
Sarkar, S., primary, Azab, B., additional, Das, S., additional, Quinn, B., additional, Shen, X., additional, Dash, R., additional, Emdad, L., additional, Thomas, S., additional, Dasgupta, S., additional, Su, Z.-Z., additional, Wang, X.-Y., additional, Sarkar, D., additional, and Fisher, P., additional
- Published
- 2013
- Full Text
- View/download PDF
12. Is there a common upstream link for autophagic and apoptotic cell death in human high-grade gliomas?
- Author
-
Emdad, L., primary, Qadeer, Z. A., additional, Bederson, L. B., additional, Kothari, H. P., additional, Uzzaman, M., additional, and Germano, I. M., additional
- Published
- 2011
- Full Text
- View/download PDF
13. Pre-clinical Experimental Therapeutics and Pharmacology
- Author
-
Jensen, R. L., primary, Gilliespie, D., additional, Ajewung, N., additional, Faure, R., additional, Kamnasaran, D., additional, Poirier, D., additional, Tamura, K., additional, Wakimoto, H., additional, Rabkin, S. D., additional, Martuza, R. L., additional, Shah, K., additional, Hashizume, R., additional, Aoki, Y., additional, Serwer, L. P., additional, Drummond, D., additional, Noble, C., additional, Park, J., additional, Bankiewicz, K., additional, James, D. C., additional, Gupta, N., additional, Agerholm-Larsen, B., additional, Iversen, H. K., additional, Jensen, K. S., additional, Moller, J., additional, Ibsen, P., additional, Mahmood, F., additional, Gehl, J., additional, Corem, E., additional, Ram, Z., additional, Daniels, D., additional, Last, D., additional, Shneor, R., additional, Salomon, S., additional, Perlstein, B., additional, Margel, S., additional, Mardor, Y., additional, Charest, G., additional, Fortin, D., additional, Mathieu, D., additional, Sanche, L., additional, Paquette, B., additional, Li, H.-F., additional, Hariono, S., additional, Dasgupta, T., additional, Kim, J.-S., additional, Haas-Kogan, D., additional, Weiss, W. A., additional, James, C. D., additional, Waldman, T., additional, Nicolaides, T., additional, Ozawa, T., additional, Rao, S., additional, Sun, H., additional, Ng, C., additional, De La Torre, J., additional, Santos, R., additional, Prados, M., additional, Butowski, N., additional, Michaud, K., additional, Solomon, D. A., additional, Prados, M. D., additional, Pandya, H., additional, Gibo, D., additional, Debinski, W., additional, Vinchon-Petit, S., additional, Jarnet, D., additional, Jadaud, E., additional, Feuvret, L., additional, Garcion, E., additional, Menei, P., additional, Chen, R., additional, Yu, J.-C., additional, Liu, C., additional, Jaffer, Z. M., additional, Chabala, J. C., additional, Winssinger, N., additional, Rubenstein, A. E., additional, Emdad, L., additional, Kothari, H., additional, Qadeer, Z., additional, Binello, E., additional, Germano, I., additional, Hirschberg, H., additional, Baek, S.-K., additional, Kwon, Y. J., additional, Sun, C. H., additional, Li, S. C., additional, Madsen, S., additional, Liu, T., additional, Wang, S.-W., additional, Gibo, D. M., additional, Fan, Q.-W., additional, Cheng, C., additional, Hackett, C., additional, Feldman, M., additional, Houseman, B. T., additional, Oakes, S. A., additional, Debnath, J., additional, Shokat, K. M., additional, Sai, K., additional, Chen, F., additional, Qiu, Z., additional, Mou, Y., additional, Zhang, X., additional, Yang, Q., additional, Chen, Z., additional, Patel, T. R., additional, Zhou, J., additional, Piepmeier, J. M., additional, Saltzman, W. M., additional, Banerjee, S., additional, Kaul, A., additional, Gianino, S. M., additional, Christians, U., additional, Gutmann, D. H., additional, Wu, J., additional, Shen, R., additional, Puduvalli, V., additional, Koul, D., additional, Alfred Yung, W. K., additional, Yun, J., additional, Sonabend, A., additional, Stuart, M., additional, Yanagihara, T., additional, Dashnaw, S., additional, Brown, T., additional, McCormick, P., additional, Romanov, A., additional, Sebastian, M., additional, Canoll, P., additional, Bruce, J. N., additional, Piao, L., additional, Joshi, K., additional, Lee, R. J., additional, Nakano, I., additional, Madsen, S. J., additional, Chou, C. C., additional, Blickenstaff, J. W., additional, Sun, C.-H., additional, Zhou, Y.-H., additional, Tome, C. M. L., additional, Wykosky, J., additional, Palma, E., additional, Nduom, E., additional, Machaidze, R., additional, Kaluzova, M., additional, Wang, Y., additional, Nie, S., additional, Hadjipanayis, C., additional, Saito, R., additional, Nakamura, T., additional, Sonoda, Y., additional, Kumabe, T., additional, Tominaga, T., additional, Lun, X., additional, Zemp, F., additional, Zhou, H., additional, Stechishin, O., additional, Kelly, J. J., additional, Weiss, S., additional, Hamilton, M. G., additional, Cairncross, G., additional, Rabinovich, B. A., additional, Bell, J., additional, McFadden, G., additional, Senger, D. L., additional, Forsyth, P. A., additional, Kang, P., additional, Jane, E. P., additional, Premkumar, D. R., additional, Pollack, I. F., additional, Yoo, J. Y., additional, Haseley, A., additional, Bratasz, A., additional, Powell, K., additional, Chiocca, E. A., additional, Kaur, B., additional, Johns, T. G., additional, Ferruzzi, P., additional, Mennillo, F., additional, De Rosa, A., additional, Rossi, M., additional, Giordano, C., additional, Magrini, R., additional, Benedetti, G., additional, Pericot, G. l., additional, Magnoni, L., additional, Mori, E., additional, Thomas, R., additional, Tunici, P., additional, Bakker, A., additional, Pradarelli, J., additional, Kaka, A., additional, Alvarez-Breckenridge, C., additional, Pan, Q., additional, Teknos, T., additional, Cen, L., additional, Ostrem, J. L., additional, Schroeder, M. A., additional, Mladek, A. C., additional, Fink, S. R., additional, Jenkins, R. B., additional, Sarkaria, J. N., additional, Madhankumar, A. B., additional, Slagle-Webb, B., additional, Park, A., additional, Pang, M., additional, Klinger, M., additional, Harbaugh, K. S., additional, Sheehan, J. M., additional, Connor, J. R., additional, Chen, T. C., additional, Wang, W., additional, Hofman, F. M., additional, Drummond, D. C., additional, Noble, C. O., additional, Park, J. W., additional, Zhou, Y., additional, Marks, J. D., additional, Alonso, M. M., additional, Gomez-Manzano, C., additional, Cortes-Santiago, N., additional, Roche, F. P., additional, Fueyo, J., additional, Johannessen, T.-C. A., additional, Grudic, A., additional, Tysnes, B. B., additional, Nigro, J., additional, Bjerkvig, R., additional, Joshi, A. D., additional, Parsons, W., additional, Velculescu, V. E., additional, Riggins, G. J., additional, Bindra, R. S., additional, Jasin, M., additional, Powell, S. N., additional, Fu, J., additional, Shen, R.-J., additional, Colman, H., additional, Lang, F. F., additional, Jensen, M. R., additional, Friedman, G. K., additional, Haas, M., additional, Cassady, K. A., additional, Gillespie, G. Y., additional, Nguyen, V., additional, Murphy, L. T., additional, Beauchamp, A. S., additional, Hollingsworth, C. K., additional, Mintz, A., additional, Garg, S., additional, Kridel, S., additional, Conrad, C. A., additional, Madden, T., additional, Ji, Y., additional, Priebe, W., additional, Seleverstov, O., additional, Purow, B. W., additional, Grant, G. A., additional, Wilson, C., additional, Campbell, M., additional, Humphries, P., additional, Li, S., additional, Li, J., additional, Johnson, A., additional, Bigner, D., additional, Dewhirst, M., additional, Pokorny, J. L., additional, Kitange, G. J., additional, Carlson, B. L., additional, Suphangul, M., additional, Petro, B., additional, Mukhtar, L., additional, Baig, M. S., additional, Villano, J., additional, Mahmud, N., additional, Keir, S. T., additional, Reardon, D. A., additional, Watson, M., additional, Shore, G. C., additional, Bigner, D. D., additional, Friedman, H. S., additional, and Gururangan, S., additional
- Published
- 2010
- Full Text
- View/download PDF
14. Melanoma differentiation associated gene-7/interleukin-24 (mda-7/IL-24): Novel gene therapeutic for metastatic melanoma
- Author
-
FISHER, P, primary, SARKAR, D, additional, LEBEDEVA, I, additional, EMDAD, L, additional, GUPTA, P, additional, SAUANE, M, additional, SU, Z, additional, GRANT, S, additional, DENT, P, additional, and CURIEL, D, additional
- Published
- 2007
- Full Text
- View/download PDF
15. Astrocyte elevated gene-1 activates cell survival pathways through PI3K-Akt signaling
- Author
-
Lee, S-G, primary, Su, Z-Z, additional, Emdad, L, additional, Sarkar, D, additional, Franke, T F, additional, and Fisher, P B, additional
- Published
- 2007
- Full Text
- View/download PDF
16. mda-7/IL-24: Multifunctional cancer-specific apoptosis-inducing cytokine
- Author
-
GUPTA, P, primary, SU, Z, additional, LEBEDEVA, I, additional, SARKAR, D, additional, SAUANE, M, additional, EMDAD, L, additional, BACHELOR, M, additional, GRANT, S, additional, CURIEL, D, additional, and DENT, P, additional
- Published
- 2006
- Full Text
- View/download PDF
17. Ionizing radiation enhances therapeutic activity of mda-7/IL-24: overcoming radiation- and mda-7/IL-24-resistance in prostate cancer cells overexpressing the antiapoptotic proteins bcl-xL or bcl-2
- Author
-
Su, Z-Z, primary, Lebedeva, I V, additional, Sarkar, D, additional, Emdad, L, additional, Gupta, P, additional, Kitada, S, additional, Dent, P, additional, Reed, J C, additional, and Fisher, P B, additional
- Published
- 2005
- Full Text
- View/download PDF
18. Ionizing radiation enhances therapeutic activity of mda-7/IL-24: overcoming radiation- and mda-7/IL-24-resistance in prostate cancer cells overexpressing the antiapoptotic proteins bcl-xL or bcl-2.
- Author
-
Su, Z.-Z., Lebedeva, I. V., Sarkar, D., Emdad, L., Gupta, P., Kitada, S., Dent, P., Reed, J. C., and Fisher, P. B.
- Subjects
CANCER cells ,CYTOKINES ,APOPTOSIS ,XENOGRAFTS ,NEOMYCIN ,PROSTATE cancer - Abstract
Subtraction hybridization applied to terminally differentiating human melanoma cells identified mda-7/IL-24, a cytokine belonging to the IL-10 gene superfamily. Adenoviral-mediated delivery of mda-7/IL-24 (Ad.mda-7) provokes apoptosis selectively in a wide spectrum of cancers in vitro in cell culture, in vivo in human tumor xenograft animal models and in patients with advanced carcinomas and melanomas. In human prostate cancer cells, a role for mitochondrial dysfunction and induction of reactive oxygen species in the apoptotic process has been established. Ectopic overexpression of bcl-x
L and bcl-2 prevents these changes including apoptosis induction in prostate tumor cells by Ad.mda-7. We now document that this resistance to apoptosis can be reversed by treating bcl-2 family overexpressing prostate tumor cells with ionizing radiation in combination with Ad.mda-7 or purified GST-MDA-7 protein. Additionally, radiation augments apoptosis induction by mda-7/IL-24 in parental and neomycin-resistant prostate tumor cells. Radiosensitization to mda-7/IL-24 is dependent on JNK signaling, as treatment with the JNK 1/2/3 inhibitor SP600125 abolishes this effect. Considering that elevated expression of bcl-xL and bcl-2 are frequent events in prostate cancer development and progression, the present studies support the use of ionizing radiation in combination with mda-7/IL-24 as a means of augmenting the therapeutic benefit of this gene in prostate cancer, particularly in the context of tumors displaying resistance to radiation therapy owing to bcl-2 family member overexpression.Oncogene (2006) 25, 2339–2348. doi:10.1038/sj.onc.1209271; published online 5 December 2005 [ABSTRACT FROM AUTHOR]- Published
- 2006
- Full Text
- View/download PDF
19. Therapy of prostate cancer using a novel cancer terminator virus and a small molecule BH-3 mimetic
- Author
-
Sarkar, S., Quinn, B. A., Shen, X. -N, rupesh dash, Das, S. K., Emdad, L., Klibanov, A. L., Wang, X. -Y, Pellecchia, M., Sarkar, D., and Fisher, P. B.
20. Transcriptional regulation of HSPB1 by Friend leukemia integration-1 factor modulates radiation and temozolomide resistance in glioblastoma
- Author
-
Yetirajam Rajesh, Biswas, A., Banik, P., Pal, I., Das, S., Borkar, S. A., Sardana, H., Saha, A., Das, S. K., Emdad, L., Fisher, P. B., and Mandal, M.
21. Retraction: mda-9/Syntenin Regulates the Metastatic Phenotype in Human Melanoma Cells by Activating Nuclear Factor-κB.
- Author
-
Boukerche H, Su ZZ, Emdad L, Sarkar D, and Fisher PB
- Published
- 2024
- Full Text
- View/download PDF
22. Noninvasive therapy of brain cancer using a unique systemic delivery methodology with a cancer terminator virus.
- Author
-
Bhoopathi P, Mannangatti P, Pradhan AK, Kumar A, Maji S, Lang FF, Klibanov AL, Madan E, Cavenee WK, Keoprasert T, Sun D, Bjerkvig R, Thorsen F, Gogna R, Das SK, Emdad L, and Fisher PB
- Subjects
- Animals, Humans, Interleukins genetics, Cell Line, Tumor, Microbubbles therapeutic use, Mice, Glioblastoma therapy, Glioblastoma virology, Glioblastoma pathology, Xenograft Model Antitumor Assays, Oncolytic Virotherapy methods, Genetic Vectors administration & dosage, Temozolomide therapeutic use, Mice, Nude, Brain Neoplasms therapy, Brain Neoplasms pathology, Brain Neoplasms virology, Blood-Brain Barrier, Adenoviridae genetics
- Abstract
Primary, glioblastoma, and secondary brain tumors, from metastases outside the brain, are among the most aggressive and therapeutically resistant cancers. A physiological barrier protecting the brain, the blood-brain barrier (BBB), functions as a deterrent to effective therapies. To enhance cancer therapy, we developed a cancer terminator virus (CTV), a unique tropism-modified adenovirus consisting of serotype 3 fiber knob on an otherwise Ad5 capsid that replicates in a cancer-selective manner and simultaneously produces a potent therapeutic cytokine, melanoma differentiation-associated gene-7/interleukin-24 (MDA-7/IL-24). A limitation of the CTV and most other viruses, including adenoviruses, is an inability to deliver systemically to treat brain tumors because of the BBB, nonspecific virus trapping, and immune clearance. These obstacles to effective viral therapy of brain cancer have now been overcome using focused ultrasound with a dual microbubble treatment, the focused ultrasound-double microbubble (FUS-DMB) approach. Proof-of-principle is now provided indicating that the BBB can be safely and transiently opened, and the CTV can then be administered in a second set of complement-treated microbubbles and released in the brain using focused ultrasound. Moreover, the FUS-DMB can be used to deliver the CTV multiple times in animals with glioblastoma growing in their brain thereby resulting in a further enhancement in survival. This strategy permits efficient therapy of primary and secondary brain tumors enhancing animal survival without promoting harmful toxic or behavioral side effects. Additionally, when combined with a standard of care therapy, Temozolomide, a further increase in survival is achieved. The FUS-DMB approach with the CTV highlights a noninvasive strategy to treat brain cancers without surgery. This innovative delivery scheme combined with the therapeutic efficacy of the CTV provides a novel potential translational therapeutic approach for brain cancers., (© 2024 Wiley Periodicals LLC.)
- Published
- 2024
- Full Text
- View/download PDF
23. Recent advances and progress in immunotherapy of solid cancers.
- Author
-
Kumar A, Emdad L, Das SK, and Fisher PB
- Subjects
- Humans, Animals, Receptors, Chimeric Antigen immunology, Immunotherapy methods, Killer Cells, Natural immunology, Neoplasms therapy, Neoplasms immunology, Immunotherapy, Adoptive methods, Tumor Microenvironment immunology
- Abstract
Adoptive cell therapy using chimeric antigen receptor (CAR) technology has become mainstream by employing advanced engineering platforms to promote cancer immunotherapy. CAR T cells have shown remarkable efficacy in the treatment of hematological malignancies; however, the value of this therapy remains inconclusive in the context of solid tumors. Immunotherapy of solid tumors is restrained by several obstacles including the presence of an immunosuppressive tumor microenvironment (TME), limited tumor trafficking, inhibited immune cell infiltration, absence of tumor-specific antigens, and off-target toxicity and adverse events associated with these therapies. Despite recent advances in CAR T cell construction, including the integration of co-stimulatory domains and the creation of armed CAR T cells, with promising outcomes in the treatment of some solid tumors, there are still many unresolved obstacles that need to be overcome. To surmount these impediments to effective CAR T cell therapies, other immune cells, such as natural killer cells and macrophages, have been engineered to serve as appealing alternatives for successful cancer immunotherapy of solid tumors. CAR NK cells demonstrate significant clinical advantages due to their ready availability and minimal toxicity. CAR macrophage (M) cells provide considerable therapeutic potential due to their ability to penetrate the TME of solid tumors. In this review, we comprehensively examine the latest developments and prospects of engineered immune cell-based cancer immunotherapies specifically designed for treating solid tumors. In addition, we provide a concise overview of current clinical trials that are examining the safety and effectiveness of modified immune cells, such as CAR T, CAR NK, and CAR M, in their ability to specifically target solid tumors and promote improved therapeutic outcomes in patients with diverse solid cancers., (Copyright © 2024. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
24. Precision medicine focus on the central nervous system: Non-invasive therapeutic agent delivery with focused ultrasound and microbubbles.
- Author
-
Mannangatti P, Bhoopathi P, Kumar A, Das SK, Emdad L, and Fisher PB
- Subjects
- Humans, Animals, Central Nervous System metabolism, Central Nervous System diagnostic imaging, Central Nervous System Diseases drug therapy, Central Nervous System Diseases therapy, Central Nervous System Diseases diagnostic imaging, Microbubbles therapeutic use, Precision Medicine methods, Drug Delivery Systems methods, Blood-Brain Barrier metabolism
- Abstract
Focused ultrasound (FUS) combined with microbubble (MB) treatment is a promising strategy capable of accurately delivering molecular medicines and gene therapy to treat various disease states. The rapid progression and use of FUS technology, from its inception to applications in contemporary medicine, exemplifies the significance and expanding potential of this technology. FUS for drug delivery in the brain can overcome challenging obstacles posed by the blood-brain barrier (BBB) in treating central nervous system (CNS) disorders. Both FUS and magnetic resonance imaging-guided FUS are non-invasive techniques for effectively opening the BBB and enhancing the transportation of molecular medicines and imaging agents into the brain. By integrating MBs into this process, it is possible to disrupt the BBB, facilitating delivery of therapeutic compounds including neuropeptides, proteins, antibodies, chemotherapeutic drugs and recently viruses accurately into the CNS. The safety and versatility of ultrasound makes it an attractive approach for administering molecular medicines, with potential applications extending beyond neurological disorders to include cancer treatment and other medical fields. Preclinical and clinical studies confirm that FUS is safe and efficient in enhancing drug administration, particularly where delivery to a precise location in the CNS is required. Combination therapies that utilize FUS and MBs also provide synergistic responses in cancer therapy. Further refining FUS and MB approaches both from a mechanical and reagent perspective will be forthcoming in the future and prove valuable in precisely defining targets and broadening therapeutic applications. Continued development and applications of FUS and MB technologies will improve therapeutic outcomes and advance patient care in multiple diseases states. This will elevate FUS and MBs from infrequently used medical options to mainstream medical applications., (Copyright © 2024. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
25. Molecular landscape of prostate cancer bone metastasis.
- Author
-
Maji S, Kumar A, Emdad L, Fisher PB, and Das SK
- Subjects
- Humans, Male, Animals, Tumor Microenvironment, Signal Transduction, Bone Neoplasms secondary, Bone Neoplasms pathology, Bone Neoplasms metabolism, Prostatic Neoplasms pathology, Prostatic Neoplasms genetics
- Abstract
Prostate cancer (PC) has a high propensity to develop bone metastases, causing severe pain and pathological fractures that profoundly impact a patients' normal functions. Current clinical intervention is mainly palliative focused on pain management, and tumor progression is refractory to standard therapeutic regimens. This limited treatment efficacy is at least partially due to a lack of comprehensive understanding of the molecular landscape of the disease pathology, along with the intensive overlapping of physiological and pathological molecular signaling. The niche is overwhelmed with diverse cell types with inter- and intra-heterogeneity, along with growth factor-enriched cells that are supportive of invading cell proliferation, providing an additional layer of complexity. This review seeks to provide molecular insights into mechanisms underlying PC bone metastasis development and progression., (Copyright © 2024. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
26. MDA-9/Syntenin in the tumor and microenvironment defines prostate cancer bone metastasis.
- Author
-
Maji S, Pradhan AK, Kumar A, Bhoopathi P, Mannangatti P, Guo C, Windle JJ, Subler MA, Wang XY, Semmes OJ, Nyalwidhe JO, Mukhopadhyay N, Paul AK, Hatfield B, Levit MM, Madan E, Sarkar D, Emdad L, Cohen DJ, Gogna R, Cavenee WK, Das SK, and Fisher PB
- Subjects
- Male, Humans, Syntenins genetics, Syntenins metabolism, Signal Transduction genetics, Cell Line, Tumor, Tumor Microenvironment, Neoplasm Metastasis, Melanoma metabolism, Prostatic Neoplasms genetics, Bone Neoplasms genetics
- Abstract
Bone metastasis is a frequent and incurable consequence of advanced prostate cancer (PC). An interplay between disseminated tumor cells and heterogeneous bone resident cells in the metastatic niche initiates this process. Melanoma differentiation associated gene-9 ( mda-9/Syntenin/syndecan binding protein ) is a prometastatic gene expressed in multiple organs, including bone marrow-derived mesenchymal stromal cells (BM-MSCs), under both physiological and pathological conditions. We demonstrate that PDGF-AA secreted by tumor cells induces CXCL5 expression in BM-MSCs by suppressing MDA-9-dependent YAP/MST signaling. CXCL5-derived tumor cell proliferation and immune suppression are consequences of the MDA-9/CXCL5 signaling axis, promoting PC disease progression. mda-9 knockout tumor cells express less PDGF-AA and do not develop bone metastases. Our data document a previously undefined role of MDA-9/Syntenin in the tumor and microenvironment in regulating PC bone metastasis. This study provides a framework for translational strategies to ameliorate health complications and morbidity associated with advanced PC.
- Published
- 2023
- Full Text
- View/download PDF
27. Cytoplasmic-delivery of polyinosine-polycytidylic acid inhibits pancreatic cancer progression increasing survival by activating Stat1-CCL2-mediated immunity.
- Author
-
Bhoopathi P, Kumar A, Pradhan AK, Maji S, Mannangatti P, Windle JJ, Subler MA, Zhang D, Vudatha V, Trevino JG, Madan E, Atfi A, Sarkar D, Gogna R, Das SK, Emdad L, and Fisher PB
- Subjects
- Animals, Humans, Mice, Chemokine CCL2 metabolism, Chemokine CCL2 therapeutic use, Cytoplasm metabolism, Cytoplasm pathology, Mice, Transgenic, Poly C therapeutic use, STAT1 Transcription Factor metabolism, Tumor Microenvironment, Carcinoma, Pancreatic Ductal genetics, Pancreatic Neoplasms metabolism
- Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer without effective therapies and with poor prognosis, causing 7% of all cancer-related fatalities in the USA. Considering the lack of effective therapies for this aggressive cancer, there is an urgent need to define newer and more effective therapeutic strategies. Polyinosine-polycytidylic acid (pIC) is a synthetic double-stranded RNA (dsRNA) which directly activates dendritic cells and natural killer cells inhibiting tumor growth. When pIC is delivered into the cytoplasm using polyethyleneimine (PEI), pIC-PEI, programmed-cell death is induced in PDAC. Transfection of [pIC]
PEI into PDAC cells inhibits growth, promotes toxic autophagy and also induces apoptosis in vitro and in vivo in animal models., Methods: The KPC transgenic mouse model that recapitulates PDAC development in patients was used to interrogate the role of an intact immune system in vivo in PDAC in response to [pIC]PEI . Antitumor efficacy and survival were monitored endpoints. Comprehensive analysis of the tumor microenvironment (TME) and immune cells, cytokines and chemokines in the spleen, and macrophage polarization were analyzed., Results: Cytosolic delivery of [pIC]PEI induces apoptosis and provokes strong antitumor immunity in vivo in immune competent mice with PDAC. The mechanism underlying the immune stimulatory properties of [pIC]PEI involves Stat1 activation resulting in CCL2 and MMP13 stimulation thereby provoking macrophage polarization. [pIC]PEI induces apoptosis via the AKT-XIAP pathway, as well as macrophage differentiation and T-cell activation via the IFNγ-Stat1-CCL2 signaling pathways in PDAC. In transgenic tumor mouse models, [pIC]PEI promotes robust and profound antitumor activity implying that stimulating the immune system contributes to biological activity. The [pIC]PEI anti-PDAC effects are enhanced when used in combination with a standard of care (SOC) treatment, that is, gemcitabine., Conclusions: In summary, [pIC]PEI treatment is non-toxic toward normal pancreatic cells while displaying strong cytotoxic and potent immune activating activities in PDAC, making it an attractive therapeutic when used alone or in conjunction with SOC therapeutic agents, potentially providing a safe and effective treatment protocol with translational potential for the effective therapy of PDAC., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2023
- Full Text
- View/download PDF
28. Dual Targeting of the PDZ1 and PDZ2 Domains of MDA-9/Syntenin Inhibits Melanoma Metastasis.
- Author
-
Pradhan AK, Modi J, Maji S, Kumar A, Bhoopathi P, Mannangatti P, Guo C, Afosah DK, Mochel MC, Mukhopadhyay ND, Kirkwood JM, Wang XY, Desai UR, Sarkar D, Emdad L, Das SK, and Fisher PB
- Subjects
- Animals, Humans, Syntenins chemistry, Signal Transduction, Peptides metabolism, Melanoma drug therapy, Melanoma genetics, Melanoma metabolism
- Abstract
Genome-wide gene expression analysis and animal modeling indicate that melanoma differentiation associated gene-9 (mda-9, Syntenin, Syndecan binding protein, referred to as MDA-9/Syntenin) positively regulates melanoma metastasis. The MDA-9/Syntenin protein contains two tandem PDZ domains serving as a nexus for interactions with multiple proteins that initiate transcription of metastasis-associated genes. Although targeting either PDZ domain abrogates signaling and prometastatic phenotypes, the integrity of both domains is critical for full biological function. Fragment-based drug discovery and NMR identified PDZ1i, an inhibitor of the PDZ1 domain that effectively blocks cancer invasion in vitro and in vivo in multiple experimental animal models. To maximize disruption of MDA-9/Syntenin signaling, an inhibitor has now been developed that simultaneously binds and blocks activity of both PDZ domains. PDZ1i was joined to the second PDZ binding peptide (TNYYFV) with a PEG linker, resulting in PDZ1i/2i (IVMT-Rx-3) that engages both PDZ domains of MDA-9/Syntenin. IVMT-Rx-3 blocks MDA-9/Syntenin interaction with Src, reduces NF-κB activation, and inhibits MMP-2/MMP-9 expression, culminating in repression of melanoma metastasis. The in vivo antimetastatic properties of IVMT-Rx-3 are enhanced when combined with an immune-checkpoint inhibitor. Collectively, our results support the feasibility of engineering MDA-9 dual-PDZ inhibitors with enhanced antimetastatic activities and applications of IVMT-Rx-3 for developing novel therapeutic strategies effectively targeting melanoma and in principle, a broad spectrum of human cancers that also overexpress MDA-9/Syntenin., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
29. A novel computational predictive biological approach distinguishes Integrin β1 as a salient biomarker for breast cancer chemoresistance.
- Author
-
Das S, Kundu M, Hassan A, Parekh A, Jena BC, Mundre S, Banerjee I, Yetirajam R, Das CK, Pradhan AK, Das SK, Emdad L, Mitra P, Fisher PB, and Mandal M
- Subjects
- Female, Humans, Biomarkers, Cell Line, Tumor, Drug Resistance, Neoplasm, Focal Adhesion Protein-Tyrosine Kinases metabolism, Breast Neoplasms drug therapy, Integrin beta1 metabolism
- Abstract
Chemoresistance is a primary cause of breast cancer treatment failure, and protein-protein interactions significantly contribute to chemoresistance during different stages of breast cancer progression. In pursuit of novel biomarkers and relevant protein-protein interactions occurring during the emergence of breast cancer chemoresistance, we used a computational predictive biological (CPB) approach. CPB identified associations of adhesion molecules with proteins connected with different breast cancer proteins associated with chemoresistance. This approach identified an association of Integrin β1 (ITGB1) with chemoresistance and breast cancer stem cell markers. ITGB1 activated the Focal Adhesion Kinase (FAK) pathway promoting invasion, migration, and chemoresistance in breast cancer by upregulating Erk phosphorylation. FAK also activated Wnt/Sox2 signaling, which enhanced self-renewal in breast cancer. Activation of the FAK pathway by ITGB1 represents a novel mechanism linked to breast cancer chemoresistance, which may lead to novel therapies capable of blocking breast cancer progression by intervening in ITGB1-regulated signaling pathways., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
30. Retraction Notice to: Insulin-like Growth Factor-binding Protein-7 (IGFBP7): A Promising Gene Therapeutic for Hepatocellular Carcinoma (HCC).
- Author
-
Chen D, Siddiq A, Emdad L, Rajasekaran D, Gredler R, Shen XN, Santhekadur PK, Srivastava J, Robertson CL, Dmitriev I, Kashentseva EA, Curiel DT, Fisher PB, and Sarkar D
- Published
- 2023
- Full Text
- View/download PDF
31. Chemoresistance in pancreatic ductal adenocarcinoma: Overcoming resistance to therapy.
- Author
-
Bhoopathi P, Mannangatti P, Das SK, Fisher PB, and Emdad L
- Subjects
- Humans, Gemcitabine, Deoxycytidine pharmacology, Deoxycytidine therapeutic use, Drug Resistance, Neoplasm, Cell Line, Tumor, Tumor Microenvironment, Pancreatic Neoplasms, Carcinoma, Pancreatic Ductal drug therapy, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal metabolism, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms metabolism
- Abstract
Pancreatic ductal adenocarcinoma (PDAC), a prominent cause of cancer deaths worldwide, is a highly aggressive cancer most frequently detected at an advanced stage that limits treatment options to systemic chemotherapy, which has provided only marginal positive clinical outcomes. More than 90% of patients with PDAC die within a year of being diagnosed. PDAC is increasing at a rate of 0.5-1.0% per year, and it is expected to be the second leading cause of cancer-related mortality by 2030. The resistance of tumor cells to chemotherapeutic drugs, which can be innate or acquired, is the primary factor contributing to the ineffectiveness of cancer treatments. Although many PDAC patients initially responds to standard of care (SOC) drugs they soon develop resistance caused partly by the substantial cellular heterogeneity seen in PDAC tissue and the tumor microenvironment (TME), which are considered key factors contributing to resistance to therapy. A deeper understanding of molecular mechanisms involved in PDAC progression and metastasis development, and the interplay of the TME in all these processes is essential to better comprehend the etiology and pathobiology of chemoresistance observed in PDAC. Recent research has recognized new therapeutic targets ushering in the development of innovative combinatorial therapies as well as enhancing our comprehension of several different cell death pathways. These approaches facilitate the lowering of the therapeutic threshold; however, the possibility of subsequent resistance development still remains a key issue and concern. Discoveries, that can target PDAC resistance, either alone or in combination, have the potential to serve as the foundation for future treatments that are effective without posing undue health risks. In this chapter, we discuss potential causes of PDAC chemoresistance and approaches for combating chemoresistance by targeting different pathways and different cellular functions associated with and mediating resistance., Competing Interests: Conflict of interest P.B.F. is a co-founder and has equity in InVaMet Therapeutics Inc. (IVMT). VCU and the Sanford Burnham Prebys Medical Discovery Institute in La Jolla, CA also have equity in IVM. SKD is the PI of a sponsored research agreement with IVMT, which is being managed by VCU., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
32. Preface.
- Author
-
Emdad L, Atfi A, Gogna R, Trevino JG, and Fisher PB
- Published
- 2023
- Full Text
- View/download PDF
33. Targeting epigenetic regulation for cancer therapy using small molecule inhibitors.
- Author
-
Kumar A, Emdad L, Fisher PB, and Das SK
- Subjects
- Humans, Chromatin, DNA Methylation, Epigenesis, Genetic, Histone Deacetylase Inhibitors therapeutic use, Histones metabolism, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Neoplasms drug therapy, Neoplasms genetics
- Abstract
Cancer cells display pervasive changes in DNA methylation, disrupted patterns of histone posttranslational modification, chromatin composition or organization and regulatory element activities that alter normal programs of gene expression. It is becoming increasingly clear that disturbances in the epigenome are hallmarks of cancer, which are targetable and represent attractive starting points for drug creation. Remarkable progress has been made in the past decades in discovering and developing epigenetic-based small molecule inhibitors. Recently, epigenetic-targeted agents in hematologic malignancies and solid tumors have been identified and these agents are either in current clinical trials or approved for treatment. However, epigenetic drug applications face many challenges, including low selectivity, poor bioavailability, instability and acquired drug resistance. New multidisciplinary approaches are being designed to overcome these limitations, e.g., applications of machine learning, drug repurposing, high throughput virtual screening technologies, to identify selective compounds with improved stability and better bioavailability. We provide an overview of the key proteins that mediate epigenetic regulation that encompass histone and DNA modifications and discuss effector proteins that affect the organization of chromatin structure and function as well as presently available inhibitors as potential drugs. Current anticancer small-molecule inhibitors targeting epigenetic modified enzymes that have been approved by therapeutic regulatory authorities across the world are highlighted. Many of these are in different stages of clinical evaluation. We also assess emerging strategies for combinatorial approaches of epigenetic drugs with immunotherapy, standard chemotherapy or other classes of agents and advances in the design of novel epigenetic therapies., Competing Interests: Conflict of interest P.B. Fisher is co-founder and has an ownership interest in InterLeukin Combinatorial Therapies (ILCT). VCU also has ownership interest in ILCT. P.B. Fisher is co-founder and has ownership interest in InVaMet Therapeutics (IVMT). VCU and the Sanford-Burnham-Prebys Medical Discovery Institute in La Jolla, CA have ownership interest in IVMT. None of the other investigators have conflicts of interest to report., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
34. Applications of tissue-specific and cancer-selective gene promoters for cancer diagnosis and therapy.
- Author
-
Kumar A, Das SK, Emdad L, and Fisher PB
- Subjects
- Humans, Oncogenes, Immunotherapy, Epigenomics, Neoplasms diagnosis, Neoplasms genetics, Neoplasms therapy, Drug-Related Side Effects and Adverse Reactions
- Abstract
Current treatment of solid tumors with standard of care chemotherapies, radiation therapy and/or immunotherapies are often limited by severe adverse toxic effects, resulting in a narrow therapeutic index. Cancer gene therapy represents a targeted approach that in principle could significantly reduce undesirable side effects in normal tissues while significantly inhibiting tumor growth and progression. To be effective, this strategy requires a clear understanding of the molecular biology of cancer development and evolution and developing biological vectors that can serve as vehicles to target cancer cells. The advent and fine tuning of omics technologies that permit the collective and spatial recognition of genes (genomics), mRNAs (transcriptomics), proteins (proteomics), metabolites (metabolomics), epiomics (epigenomics, epitranscriptomics, and epiproteomics), and their interactomics in defined complex biological samples provide a roadmap for identifying crucial targets of relevance to the cancer paradigm. Combining these strategies with identified genetic elements that control target gene expression uncovers significant opportunities for developing guided gene-based therapeutics for cancer. The purpose of this review is to overview the current state and potential limitations in developing gene promoter-directed targeted expression of key genes and highlights their potential applications in cancer gene therapy., (Copyright © 2023. Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
35. Enhanced Cancer Therapy Using an Engineered Designer Cytokine Alone and in Combination With an Immune Checkpoint Inhibitor.
- Author
-
Pradhan AK, Bhoopathi P, Maji S, Kumar A, Guo C, Mannangatti P, Li J, Wang XY, Sarkar D, Emdad L, Das SK, and Fisher PB
- Abstract
melanoma differentiation associated gene-7 or Interleukin-24 ( mda-7, IL-24 ) displays expansive anti-tumor activity without harming corresponding normal cells/tissues. This anticancer activity has been documented in vitro and in vivo in multiple preclinical animal models, as well as in patients with advanced cancers in a phase I clinical trial. To enhance the therapeutic efficacy of MDA-7 (IL-24), we engineered a designer cytokine (a "Superkine"; IL-24S; referred to as M7S) with enhanced secretion and increased stability to engender improved "bystander" antitumor effects. M7S was engineered in a two-step process by first replacing the endogenous secretory motif with an alternate secretory motif to boost secretion. Among four different signaling peptides, the insulin secretory motif significantly enhanced the secretion of MDA-7 (IL-24) protein and was chosen for M7S. The second modification engineered in M7S was designed to enhance the stability of MDA-7 (IL-24), which was accomplished by replacing lysine at position K122 with arginine. This engineered "M7S Superkine" with increased secretion and stability retained cancer specificity. Compared to parental MDA-7 (IL-24), M7S (IL-24S) was superior in promoting anti-tumor and bystander effects leading to improved outcomes in multiple cancer xenograft models. Additionally, combinatorial therapy using MDA-7 (IL-24) or M7S (IL-24S) with an immune checkpoint inhibitor, anti-PD-L1, dramatically reduced tumor progression in murine B16 melanoma cells. These results portend that M7S (IL-24S) promotes the re-emergence of an immunosuppressive tumor microenvironment, providing a solid rationale for prospective translational applications of this therapeutic designer cytokine., Competing Interests: PBF is a co-founder and has equity in InterLeukin Combinatorial Therapies, Inc. (ILCT). VCU also has equity in ILCT. LE is the PI of a sponsored research agreement with ILCT, which is being managed by VCU. The remaining 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. The authors declare that this study received funding from InterLeukin Combinatorial Therapies (ILCT), Inc. ILCT supported IL-24-related research through a sponsored research agreement between ILCT and VCU School of Medicine., (Copyright © 2022 Pradhan, Bhoopathi, Maji, Kumar, Guo, Mannangatti, Li, Wang, Sarkar, Emdad, Das and Fisher.)
- Published
- 2022
- Full Text
- View/download PDF
36. Conversion of a Non-Cancer-Selective Promoter into a Cancer-Selective Promoter.
- Author
-
Bhoopathi P, Pradhan AK, Kumar A, Maji S, Mannangatti P, Deng X, Bandyopadhyay D, Sarkar D, Wang XY, Landry JW, Das SK, Emdad L, and Fisher PB
- Abstract
Progression-elevated gene-3 (PEG-3) and rat growth arrest and DNA damage-inducible gene-34 (GADD34) display significant sequence homology with regulation predominantly transcriptional. The rat full-length (FL) and minimal (min) PEG-3 promoter display cancer-selective expression in rodent and human tumors, allowing for cancer-directed regulation of transgenes, viral replication and in vivo imaging of tumors and metastases in animals, whereas the FL- and min-GADD34-Prom lack cancer specificity. Min-PEG-Prom and min-GADD34-Prom have identical sequences except for two single-point mutation differences (at -260 bp and +159 bp). Engineering double mutations in the min-GADD34-Prom produce the GAPE-Prom. Changing one base pair (+159) or both point mutations in the min-GADD34-Prom, but not the FL-GADD34-Prom, results in cancer-selective transgene expression in diverse cancer cells (including prostate, breast, pancreatic and neuroblastoma) vs. normal counterparts. Additionally, we identified a GATA2 transcription factor binding site, promoting cancer specificity when both min-PEG-Prom mutations are present in the GAPE-Prom. Taken together, introducing specific point mutations in a rat min-GADD34-Prom converts this non-cancer-specific promoter into a cancer-selective promoter, and the addition of GATA2 with existing AP1 and PEA3 transcription factors enhances further cancer-selective activity of the GAPE-Prom. The GAPE-Prom provides a genetic tool to specifically regulate transgene expression in cancer cells.
- Published
- 2022
- Full Text
- View/download PDF
37. Screening of the Prime bioactive compounds from Aloe vera as potential anti-proliferative agents targeting DNA.
- Author
-
Majumder R, Das CK, Banerjee I, Chandra Jena B, Mandal A, Das P, Pradhan AK, Das S, Basak P, Das SK, Emdad L, Fisher PB, and Mandal M
- Subjects
- DNA, Humans, Molecular Docking Simulation, Plant Extracts pharmacology, Aloe chemistry, Neoplasms
- Abstract
Background: Aloe vera extract and its bioactive compounds possess anti-proliferative properties against cancer cells. However, no detailed molecular mechanism of action studies has been reported. We have now employed a computational approach to scrutinize the molecular mechanism of lead bioactive compounds from Aloe vera that potentially inhibit DNA synthesis., Methods: Initially, the anti-proliferative activity of Aloe vera extract was examined in human breast cancer cells (in vitro/in vivo). Later on, computational screening of bioactive compounds from Aloe vera targeting DNA was performed by molecular docking and molecular dynamics simulation., Results: In-vitro and in-vivo studies confirm that Aloe vera extract effectively suppresses the growth of breast cancer cells without significant cytotoxicity towards non-cancerous normal immortal cells. Computational screening predicts that growth suppression may be due to the presence of DNA intercalating bioactive compounds (riboflavin, daidzin, aloin, etc.) contained in Aloe vera. MM/PBSA calculation showed that riboflavin has a higher binding affinity at the DNA binding sites compared to standard drug daunorubicin., Conclusions: These observations support the hypothesis that riboflavin may be exploited as an anti-proliferative DNA intercalating agent to prevent cancer and is worthy of testing for the management of cancer by performing more extensive pre-clinical and if validated clinical trials., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
38. GAP junctions: multifaceted regulators of neuronal differentiation.
- Author
-
Talukdar S, Emdad L, Das SK, and Fisher PB
- Subjects
- Animals, Mice, Mice, Knockout, Neurons chemistry, Neurons metabolism, Connexins analysis, Connexins genetics, Connexins metabolism, Gap Junctions chemistry, Gap Junctions metabolism
- Abstract
Gap junctions are intercellular membrane channels consisting of connexin proteins, which contribute to direct cytoplasmic exchange of small molecules, substrates and metabolites between adjacent cells. These channels play important roles in neuronal differentiation, maintenance, survival and function. Gap junctions regulate differentiation of neurons from embryonic, neural and induced pluripotent stem cells. In addition, they control transdifferentiation of neurons from mesenchymal stem cells. The expression and levels of several connexins correlate with cell cycle changes and different stages of neurogenesis. Connexins such as Cx36, Cx45, and Cx26, play a crucial role in neuronal function. Several connexin knockout mice display lethal or severely impaired phenotypes. Aberrations in connexin expression is frequently associated with various neurodegenerative disorders. Gap junctions also act as promising therapeutic targets for neuronal regenerative medicine, because of their role in neural stem cell integration, injury and remyelination.
- Published
- 2022
- Full Text
- View/download PDF
39. Insights into the Mechanisms of Action of MDA-7/IL-24: A Ubiquitous Cancer-Suppressing Protein.
- Author
-
Modi J, Roy A, Pradhan AK, Kumar A, Talukdar S, Bhoopathi P, Maji S, Mannangatti P, Sanchez De La Rosa D, Li J, Guo C, Subler MA, Windle JJ, Cavenee WK, Sarkar D, Wang XY, Das SK, Emdad L, and Fisher PB
- Subjects
- Animals, Apoptosis physiology, Autophagy physiology, Cell Death physiology, Humans, Melanoma metabolism, Interleukins metabolism, Tumor Suppressor Proteins metabolism
- Abstract
Melanoma differentiation associated gene-7/interleukin-24 (MDA-7/IL-24), a secreted protein of the IL-10 family, was first identified more than two decades ago as a novel gene differentially expressed in terminally differentiating human metastatic melanoma cells. MDA-7/IL-24 functions as a potent tumor suppressor exerting a diverse array of functions including the inhibition of tumor growth, invasion, angiogenesis, and metastasis, and induction of potent "bystander" antitumor activity and synergy with conventional cancer therapeutics. MDA-7/IL-24 induces cancer-specific cell death through apoptosis or toxic autophagy, which was initially established in vitro and in preclinical animal models in vivo and later in a Phase I clinical trial in patients with advanced cancers. This review summarizes the history and our current understanding of the molecular/biological mechanisms of MDA-7/IL-24 action rendering it a potent cancer suppressor.
- Published
- 2021
- Full Text
- View/download PDF
40. SARI inhibits growth and reduces survival of oral squamous cell carcinomas (OSCC) by inducing endoplasmic reticulum stress.
- Author
-
Priyadarshini M, Maji S, Samal SK, Rath R, Li J, Das SK, Emdad L, Kundu CN, Fisher PB, and Dash R
- Subjects
- Animals, Carcinoma, Squamous Cell pathology, Cell Line, Tumor, Cell Survival physiology, HEK293 Cells, Humans, Male, Mice, Mice, Inbred BALB C, Mice, Nude, Mouth Neoplasms pathology, Squamous Cell Carcinoma of Head and Neck pathology, Xenograft Model Antitumor Assays methods, Basic-Leucine Zipper Transcription Factors biosynthesis, Carcinoma, Squamous Cell metabolism, Endoplasmic Reticulum Stress physiology, Growth Inhibitors biosynthesis, Mouth Neoplasms metabolism, Squamous Cell Carcinoma of Head and Neck metabolism, Tumor Suppressor Proteins biosynthesis
- Abstract
Aims: SARI (suppressor of activator protein (AP)-1, regulated by interferon (IFN) was identified as a novel tumor suppressor by applying subtraction hybridization to terminally differentiating human melanoma cells. The anti-tumor activity of SARI and the correlation between expression and cancer aggression and metastasis has been examined in multiple cancers, but its potential role in oral squamous cell carcinomas (OSCC) has not been explored., Methods: SARI expression was monitored in tumor tissues of OSCC patients by performing immunohistochemistry. Ectopic expression of SARI was achieved using a replication defective adenovirus expressing SARI (Ad.SARI). A nude mouse xenograft model was used to evaluate the in vivo efficacy of SARI. Endoplasmic reticulum (ER) stress was monitored in SARI infected OSCC cells by confocal microscopy., Key Finding: In this study, we demonstrate that SARI expression is significantly lower in OSCC tumor tissue as compared to normal adjacent tissue. Ectopic expression of SARI induces cancer-specific cell death in human OSCC cell lines and in a paclitaxel plus cisplatin non-responder OSCC patient-derived (PDC1) cell line. Mechanistically, SARI inhibits zinc finger protein GLI1 expression through induction of endoplasmic reticulum (ER) stress. Using a nude mouse xenograft model, we show that intratumoral injections of Ad.SARI significantly reduce PDC1 tumor burden, whereas treatment with an ER stress inhibitor efficiently rescues tumors from growth inhibition., Significance: Overall, our data provides a link between induction of ER stress and inhibition of the GLI1/Hedgehog signaling pathway and the tumor suppressive activity of SARI in the context of OSCC., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
41. The quest to develop an effective therapy for neuroblastoma.
- Author
-
Bhoopathi P, Mannangatti P, Emdad L, Das SK, and Fisher PB
- Subjects
- Animals, Antineoplastic Agents adverse effects, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Gene Expression Regulation, Neoplastic, Humans, Molecular Targeted Therapy, Neoplasm Staging, Neuroblastoma genetics, Neuroblastoma metabolism, Neuroblastoma pathology, Radiation Dosage, Signal Transduction, Antineoplastic Agents therapeutic use, Immunotherapy adverse effects, Neuroblastoma therapy
- Abstract
Neuroblastoma (NB) is a common solid extracranial tumor developing in pediatric populations. NB can spontaneously regress or grow and metastasize displaying resistance to therapy. This tumor is derived from primitive cells, mainly those of the neural crest, in the sympathetic nervous system and usually develops in the adrenal medulla and paraspinal ganglia. Our understanding of the molecular characteristics of human NBs continues to advance documenting abnormalities at the genome, epigenome, and transcriptome levels. The high-risk tumors have MYCN oncogene amplification, and the MYCN transcriptional regulator encoded by the MYCN oncogene is highly expressed in the neural crest. Studies on the biology of NB has enabled a more precise risk stratification strategy and a concomitant reduction in the required treatment in an expanding number of cases worldwide. However, newer treatment strategies are mandated to improve outcomes in pediatric patients who are at high-risk and display relapse. To improve outcomes and survival rates in such high-risk patients, it is necessary to use a multicomponent therapeutic approach. Accuracy in clinical staging of the disease and assessment of the associated risks based on biological, clinical, surgical, and pathological criteria are of paramount importance for prognosis and to effectively plan therapeutic approaches. This review discusses the staging of NB and the biological and genetic features of the disease and several current therapies including targeted delivery of chemotherapy, novel radiation therapy, and immunotherapy for NB., (© 2021 Wiley Periodicals LLC.)
- Published
- 2021
- Full Text
- View/download PDF
42. Pharmacological inhibition of MDA-9/Syntenin blocks breast cancer metastasis through suppression of IL-1β.
- Author
-
Pradhan AK, Maji S, Bhoopathi P, Talukdar S, Mannangatti P, Guo C, Wang XY, Cartagena LC, Idowu M, Landry JW, Sarkar D, Emdad L, Cavenee WK, Das SK, and Fisher PB
- Subjects
- Animals, Antineoplastic Agents chemical synthesis, Breast Neoplasms genetics, Breast Neoplasms immunology, Breast Neoplasms pathology, Cell Line, Tumor, Chemokine CCL11 genetics, Chemokine CCL11 immunology, Chemokine CCL17 genetics, Chemokine CCL17 immunology, Female, Gene Expression Regulation, Neoplastic, Humans, Interleukin-10 genetics, Interleukin-10 immunology, Interleukin-1alpha genetics, Interleukin-1alpha immunology, Interleukin-1beta antagonists & inhibitors, Interleukin-1beta immunology, Interleukin-23 Subunit p19 genetics, Interleukin-23 Subunit p19 immunology, Interleukin-5 genetics, Interleukin-5 immunology, Lung Neoplasms genetics, Lung Neoplasms immunology, Lung Neoplasms secondary, Mice, Mice, Inbred BALB C, Oxadiazoles chemical synthesis, Pyrimidines chemical synthesis, Signal Transduction, Syntenins antagonists & inhibitors, Syntenins immunology, T-Lymphocytes, Cytotoxic drug effects, T-Lymphocytes, Cytotoxic immunology, T-Lymphocytes, Cytotoxic pathology, Tumor Burden drug effects, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Breast Neoplasms drug therapy, Interleukin-1beta genetics, Lung Neoplasms drug therapy, Oxadiazoles pharmacology, Pyrimidines pharmacology, Syntenins genetics
- Abstract
Melanoma differentiation associated gene-9 (MDA-9), Syntenin-1, or syndecan binding protein is a differentially regulated prometastatic gene with elevated expression in advanced stages of melanoma. MDA-9/Syntenin expression positively associates with advanced disease stage in multiple histologically distinct cancers and negatively correlates with patient survival and response to chemotherapy. MDA-9/Syntenin is a highly conserved PDZ-domain scaffold protein, robustly expressed in a spectrum of diverse cancer cell lines and clinical samples. PDZ domains interact with a number of proteins, many of which are critical regulators of signaling cascades in cancer. Knockdown of MDA-9/Syntenin decreases cancer cell metastasis, sensitizing these cells to radiation. Genetic silencing of MDA-9/Syntenin or treatment with a pharmacological inhibitor of the PDZ1 domain, PDZ1i, also activates the immune system to kill cancer cells. Additionally, suppression of MDA-9/Syntenin deregulates myeloid-derived suppressor cell differentiation via the STAT3/interleukin (IL)-1β pathway, which concomitantly promotes activation of cytotoxic T lymphocytes. Biologically, PDZ1i treatment decreases metastatic nodule formation in the lungs, resulting in significantly fewer invasive cancer cells. In summary, our observations indicate that MDA-9/Syntenin provides a direct therapeutic target for mitigating aggressive breast cancer and a small-molecule inhibitor, PDZ1i, provides a promising reagent for inhibiting advanced breast cancer pathogenesis., Competing Interests: Competing interest statement: W.K.C. and P.B.F. are cofounders and have ownership interest in InVaMet Therapeutics, Inc. Virginia Commonwealth University and the Sanford Burnham Prebys Medical Discovery Institute have ownership interest in InVaMet Therapeutics, Inc.
- Published
- 2021
- Full Text
- View/download PDF
43. Corrigendum to "Prevention of epithelial to mesenchymal transition in colorectal carcinoma by regulation of the E-cadherin-β-catenin-vinculin axis" [Cancer Lett. 452 (2019) 254-263].
- Author
-
Pal I, Rajesh Y, Banik P, Dey G, Dey KK, Bharti R, Naskar D, Chakraborty S, Ghosh SK, Das SK, Emdad L, Chandra Kundu S, Fisher PB, and Mandal M
- Published
- 2021
- Full Text
- View/download PDF
44. Theranostic Tripartite Cancer Terminator Virus for Cancer Therapy and Imaging.
- Author
-
Bhoopathi P, Pradhan AK, Maji S, Das SK, Emdad L, and Fisher PB
- Abstract
Combining cancer-selective viral replication and simultaneous production of a therapeutic cytokine, with potent "bystander" anti-tumor activity, are hallmarks of the cancer terminator virus ( CTV ). To expand on these attributes, we designed a next generation CTV that additionally enables simultaneous non-invasive imaging of tumors targeted for eradication. A unique tripartite CTV "theranostic" adenovirus ( TCTV ) has now been created that employs three distinct promoters to target virus replication, cytokine production and imaging capabilities uniquely in cancer cells. Conditional replication of the TCTV is regulated by a cancer-selective (truncated PEG-3 ) promoter, the therapeutic component, MDA-7/IL-24, is under a ubiquitous ( CMV ) promoter, and finally the imaging capabilities are synchronized through another cancer selective (truncated tCCN1 ) promoter. Using in vitro studies and clinically relevant in vivo models of breast and prostate cancer, we demonstrate that incorporating a reporter gene for imaging does not compromise the exceptional therapeutic efficacy of our previously reported bipartite CTV . This TCTV permits targeted treatment of tumors while monitoring tumor regression, with potential to simultaneously detect metastasis due to the cancer-selective activity of reporter gene expression. This "theranostic" virus provides a new genetic tool for distinguishing and treating localized and metastatic cancers.
- Published
- 2021
- Full Text
- View/download PDF
45. Metabolic control of cancer progression as novel targets for therapy.
- Author
-
Talukdar S, Emdad L, Gogna R, Das SK, and Fisher PB
- Subjects
- Cell Transformation, Neoplastic, Humans, Metabolic Networks and Pathways, Oncogenes, Tumor Microenvironment, Neoplasms drug therapy, Neoplasms genetics
- Abstract
Metabolism is an important part of tumorigenesis as well as progression. The various cancer metabolism pathways, such as glucose metabolism and glutamine metabolism, directly regulate the development and progression of cancer. The pathways by which the cancer cells rewire their metabolism according to their needs, surrounding environment and host tissue conditions are an important area of study. The regulation of these metabolic pathways is determined by various oncogenes, tumor suppressor genes, as well as various constituent cells of the tumor microenvironment. Expanded studies on metabolism will help identify efficient biomarkers for diagnosis and strategies for therapeutic interventions and countering ways by which cancers may acquire resistance to therapy., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
46. Autophagy and senescence: Insights from normal and cancer stem cells.
- Author
-
Talukdar S, Das SK, Emdad L, and Fisher PB
- Subjects
- Aging physiology, Animals, Apoptosis physiology, Cell Differentiation physiology, Humans, Neoplastic Stem Cells pathology, Stress, Physiological physiology, Autophagy physiology, Cellular Senescence physiology, Neoplastic Stem Cells physiology, Stem Cells physiology
- Abstract
Autophagy is a fundamental cellular process, which allows cells to adapt to metabolic stress through the degradation and recycling of intracellular components to generate macromolecular precursors and produce energy. Autophagy is also critical in maintaining cellular/tissue homeostasis, as well preserving immunity and preventing human disease. Deregulation of autophagic processes is associated with cancer, neurodegeneration, muscle and heart disease, infectious diseases and aging. Research on a variety of stem cell types establish that autophagy plays critical roles in normal and cancer stem cell quiescence, activation, differentiation, and self-renewal. Considering its critical function in regulating the metabolic state of stem cells, autophagy plays a dual role in the regulation of normal and cancer stem cell senescence, and cellular responses to various therapeutic strategies. The relationships between autophagy, senescence, dormancy and apoptosis frequently focus on responses to various forms of stress. These are interrelated processes that profoundly affect normal and abnormal human physiology that require further elucidation in cancer stem cells. This review provides a current perspective on autophagy and senescence in both normal and cancer stem cells., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
47. Identification of Annexin A2 as a key mTOR target to induce roller coaster pattern of autophagy fluctuation in stress.
- Author
-
Mukhopadhyay S, Praharaj PP, Naik PP, Talukdar S, Emdad L, Das SK, Fisher PB, and Bhutia SK
- Subjects
- A549 Cells, HeLa Cells, Humans, Phosphorylation, Tumor Cells, Cultured, Annexin A2 metabolism, Autophagy, Stress, Physiological, TOR Serine-Threonine Kinases metabolism
- Abstract
Autophagy can either be cytoprotective or promote cell death in a context-dependent manner in response to stress. How autophagy leads to autophagy dependent cell death requires further clarification. In this study, we document a nonlinear roller coaster form of autophagy oscillation when cells are subjected to different stress conditions. Serum starvation induces an initial primary autophagic peak at 6 h, that helps to replenish cells with de novo fluxed nutrients, but protracted stress lead to a secondary autophagic peak around 48 h. Time kinetic studies indicate that the primary autophagic peak is reversible, whereas the secondary autophagic peak is irreversible and leads to cell death. Key players involved in different stages of autophagy including initiation, elongation and degradation during this oscillatory sequence were identified. A similar molecular pattern was intensified under apoptosis-deficient conditions. mTOR was the central molecule regulating this autophagic activity, and upon knockdown a steady increase of autophagy without any non-linear fluctuation was evident. An unbiased proteome screening approach was employed to identify the autophagy molecules potentially regulating these autophagic peaks. Our proteomics analysis has identified Annexin A2 as a stress-induced protein to implicate in autophagy fluctuation and its deficiency reduced autophagy. Moreover, we report that mTOR in its phosphorylated condition interacts with Annexin A2 to induce autophagy fluctuation by altering its cellular localization. The work highlights the molecular mechanism of a mTOR-dependent roller coaster fluctuation of autophagy and autophagy dependent cell death during prolong stress., (Copyright © 2020 Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
48. Recent insights into apoptosis and toxic autophagy: The roles of MDA-7/IL-24, a multidimensional anti-cancer therapeutic.
- Author
-
Emdad L, Bhoopathi P, Talukdar S, Pradhan AK, Sarkar D, Wang XY, Das SK, and Fisher PB
- Subjects
- Animals, Apoptosis drug effects, Autophagy drug effects, Cell Death physiology, Clinical Trials, Phase I as Topic, Clinical Trials, Phase II as Topic, Humans, Neoplasms pathology, Antineoplastic Agents pharmacology, Apoptosis physiology, Autophagy physiology, Interleukins metabolism, Neoplasms drug therapy, Neoplasms metabolism
- Abstract
Apoptosis and autophagy play seminal roles in maintaining organ homeostasis. Apoptosis represents canonical type I programmed cell death. Autophagy is viewed as pro-survival, however, excessive autophagy can promote type II cell death. Defective regulation of these two obligatory cellular pathways is linked to various diseases, including cancer. Biologic or chemotherapeutic agents, which can reprogram cancer cells to undergo apoptosis- or toxic autophagy-mediated cell death, are considered effective tools for treating cancer. Melanoma differentiation associated gene-7 (mda-7) selectively promotes these effects in cancer cells. mda-7 was identified more than two decades ago by subtraction hybridization showing elevated expression during induction of terminal differentiation of metastatic melanoma cells following treatment with recombinant fibroblast interferon and mezerein (a PKC activating agent). MDA-7 was classified as a member of the IL-10 gene family based on its chromosomal location, and the presence of an IL-10 signature motif and a secretory sequence, and re-named interleukin-24 (MDA-7/IL-24). Multiple studies have established MDA-7/IL-24 as a potent anti-cancer agent, which when administered at supra-physiological levels induces growth arrest and cell death through apoptosis and toxic autophagy in a wide variety of tumor cell types, but not in corresponding normal/non-transformed cells. Furthermore, in a phase I/II clinical trial, MDA-7/IL-24 administered by means of a non-replicating adenovirus was well tolerated and displayed significant clinical activity in patients with multiple advanced cancers. This review examines our current comprehension of the role of MDA-7/IL-24 in mediating cancer-specific cell death via apoptosis and toxic autophagy., (Copyright © 2019. Published by Elsevier Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
49. MDA-9/Syntenin/SDCBP: new insights into a unique multifunctional scaffold protein.
- Author
-
Pradhan AK, Maji S, Das SK, Emdad L, Sarkar D, and Fisher PB
- Subjects
- Animals, Humans, Neoplasm Metastasis, Neoplasms genetics, Neoplasms pathology, Syntenins genetics, Neoplasms metabolism, Syntenins metabolism
- Abstract
Tumor metastasis comprises a series of coordinated events that culminate in dissemination of cancer cells to distant sites within the body representing the greatest challenge impeding effective therapy of cancer and the leading cause of cancer-associated morbidity. Cancer cells exploit multiple genes and pathways to colonize to distant organs. These pathways are integrated and regulated at different levels by cellular- and extracellular-associated factors. Defining the genes and pathways that govern metastasis can provide new targets for therapeutic intervention. Melanoma differentiation associated gene-9 (mda-9) (also known as Syntenin-1 and SDCBP (Syndecan binding protein)) was identified by subtraction hybridization as a novel gene displaying differential temporal expression during differentiation of melanoma. MDA-9/Syntenin is an established Syndecan binding protein that functions as an adaptor protein. Expression of MDA-9/Syntenin is elevated at an RNA and protein level in a wide-range of cancers including melanoma, glioblastoma, neuroblastoma, and prostate, breast and liver cancer. Expression is increased significantly in metastatic cancer cells as compared with non-metastatic cancer cells or normal cells, which make it an attractive target in treating cancer metastasis. In this review, we focus on the role and regulation of mda-9 in cancer progression and metastasis.
- Published
- 2020
- Full Text
- View/download PDF
50. Lumefantrine, an antimalarial drug, reverses radiation and temozolomide resistance in glioblastoma.
- Author
-
Rajesh Y, Biswas A, Kumar U, Banerjee I, Das S, Maji S, Das SK, Emdad L, Cavenee WK, Mandal M, and Fisher PB
- Subjects
- Brain Neoplasms radiotherapy, Cell Line, Tumor, Drug Resistance, Neoplasm, Epithelial-Mesenchymal Transition drug effects, Gene Expression Regulation, Neoplastic drug effects, Glioblastoma genetics, Glioblastoma metabolism, Glioblastoma radiotherapy, Heat-Shock Proteins genetics, Heat-Shock Proteins metabolism, Humans, Microfilament Proteins genetics, Microfilament Proteins metabolism, Molecular Chaperones genetics, Molecular Chaperones metabolism, Trans-Activators genetics, Trans-Activators metabolism, Antimalarials administration & dosage, Antineoplastic Agents, Alkylating administration & dosage, Brain Neoplasms drug therapy, Glioblastoma drug therapy, Lumefantrine administration & dosage, Temozolomide administration & dosage
- Abstract
Glioblastoma multiforme (GBM) is an aggressive cancer without currently effective therapies. Radiation and temozolomide (radio/TMZ) resistance are major contributors to cancer recurrence and failed GBM therapy. Heat shock proteins (HSPs), through regulation of extracellular matrix (ECM) remodeling and epithelial mesenchymal transition (EMT), provide mechanistic pathways contributing to the development of GBM and radio/TMZ-resistant GBM. The Friend leukemia integration 1 (Fli-1) signaling network has been implicated in oncogenesis in GBM, making it an appealing target for advancing novel therapeutics. Fli-1 is linked to oncogenic transformation with up-regulation in radio/TMZ-resistant GBM, transcriptionally regulating HSPB1. This link led us to search for targeted molecules that inhibit Fli-1. Expression screening for Fli-1 inhibitors identified lumefantrine, an antimalarial drug, as a probable Fli-1 inhibitor. Docking and isothermal calorimetry titration confirmed interaction between lumefantrine and Fli-1. Lumefantrine promoted growth suppression and apoptosis in vitro in parental and radio/TMZ-resistant GBM and inhibited tumor growth without toxicity in vivo in U87MG GBM and radio/TMZ-resistant GBM orthotopic tumor models. These data reveal that lumefantrine, an FDA-approved drug, represents a potential GBM therapeutic that functions through inhibition of the Fli-1/HSPB1/EMT/ECM remodeling protein networks., Competing Interests: The authors declare no competing interest.
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.