1
|
Cabrera M, Armando R, Czarnowski I, Chinestrad P, Blanco R, Zinni A, Gómez D, Mengual Gómez DL, Menna PL. CADD-based discovery of novel oligomeric modulators of PKM2 with antitumor activity in aggressive human glioblastoma models. Heliyon 2025; 11:e42238. [PMID: 39959478 PMCID: PMC11830341 DOI: 10.1016/j.heliyon.2025.e42238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/22/2025] [Accepted: 01/22/2025] [Indexed: 02/18/2025] Open
Abstract
Pyruvate kinase isoform M2 (PKM2) is a multifunctional enzyme capable of transitioning between monomeric, dimeric, and tetrameric states, with its oligomeric equilibrium playing a pivotal role in tumour progression and survival. The unique exon ten at the dimer-dimer interface represents an attractive target for isoform-specific modulation, offering opportunities for disrupting this equilibrium and altering tumour cell dynamics. This study identifies a novel druggable pocket at the PKM2 dimer interface through conformational analysis. This pocket was exploited in a virtual screening of a large small-molecule library, identifying two promising candidates, C599 and C998. Both compounds exhibited dose-dependent antiproliferative effects in glioblastoma cell lines and induced apoptosis, as evidenced by caspase 3/7 activation. These effects were directly linked to their inhibition of PKM2 enzymatic activity, validating the proposed mechanism of action in their rational design. ADMET studies further highlighted their strong potential as lead PKM2 inhibitors for GBM treatment. Molecular dynamics (MD) simulations and post-MD analyses, including Dynamic Cross-Correlation Maps (DCCM), Probability Density Function (PDF), and Free Energy Landscape (FEL), confirmed the stability of the protein-ligand interactions and highlighted critical residues at the dimer-dimer interface. The Steered MD simulations demonstrated the high affinity of the compounds for PKM2, as evidenced by the requirement of high rupture forces to induce an unbinding event. These results highlight the potential of the compounds as oligomeric modulators of PKM2. These findings position C599 and C998 as promising lead compounds for antitumor applications. Future studies will focus on optimising these candidates and assessing their efficacy in vivo glioblastoma models, reassuring the thoroughness of our research and the potential for further advancements.
Collapse
Affiliation(s)
- Maia Cabrera
- Laboratorio de Farmacología Molecular, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Argentina
| | - Romina Armando
- Unidad de Oncología Molecular, Centro de Oncología Molecular y Traslacional, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Argentina
| | - Ian Czarnowski
- Laboratorio de Farmacología Molecular, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Argentina
| | - Patricio Chinestrad
- Laboratorio de Farmacología Molecular, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Argentina
| | - Ramiro Blanco
- Laboratorio de Farmacología Molecular, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Argentina
| | - Alejandra Zinni
- Laboratorio de Farmacología Molecular, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Argentina
| | - Daniel Gómez
- Unidad de Oncología Molecular, Centro de Oncología Molecular y Traslacional, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Argentina
| | - Diego L. Mengual Gómez
- Unidad de Oncología Molecular, Centro de Oncología Molecular y Traslacional, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Argentina
| | - Pablo Lorenzano Menna
- Laboratorio de Farmacología Molecular, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Argentina
| |
Collapse
|
2
|
Vázquez Cervantes GI, González Esquivel DF, Gómez-Manzo S, Pineda B, Pérez de la Cruz V. New Immunotherapeutic Approaches for Glioblastoma. J Immunol Res 2021; 2021:3412906. [PMID: 34557553 PMCID: PMC8455182 DOI: 10.1155/2021/3412906] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/24/2021] [Indexed: 12/27/2022] Open
Abstract
Glioblastoma (GBM) is the most common primary malignant brain tumor with a high mortality rate. The current treatment consists of surgical resection, radiation, and chemotherapy; however, the median survival rate is only 12-18 months despite these alternatives, highlighting the urgent need to find new strategies. The heterogeneity of GBM makes this tumor difficult to treat, and the immunotherapies result in an attractive approach to modulate the antitumoral immune responses favoring the tumor eradication. The immunotherapies for GMB including monoclonal antibodies, checkpoint inhibitors, vaccines, and oncolytic viruses, among others, have shown favorable results alone or as a multimodal treatment. In this review, we summarize and discuss promising immunotherapies for GBM currently under preclinical investigation as well as in clinical trials.
Collapse
Affiliation(s)
- Gustavo Ignacio Vázquez Cervantes
- Neurochemistry and Behavior Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Edificio A, 1° Piso, Circuito de Posgrados, Ciudad Universitaria, Coyoacán, C.P. 04510 Distrito Federal, Mexico
| | - Dinora F. González Esquivel
- Neurochemistry and Behavior Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico
| | - Saúl Gómez-Manzo
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, México City 04530, Mexico
| | - Benjamín Pineda
- Neuroimmunology Department, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico
| | - Verónica Pérez de la Cruz
- Neurochemistry and Behavior Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico
| |
Collapse
|
3
|
Rana A, Bhatnagar S. Advancements in folate receptor targeting for anti-cancer therapy: A small molecule-drug conjugate approach. Bioorg Chem 2021; 112:104946. [PMID: 33989916 DOI: 10.1016/j.bioorg.2021.104946] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/17/2021] [Accepted: 04/22/2021] [Indexed: 10/21/2022]
Abstract
Targeted delivery combined with controlled release of drugs has a crucial role in future of personalized medicine. The majority of cancer drugs are intended to interfere with one or more cellular events. Anticancer agents can also be toxic to healthy cells, as healthy cells may also need to proliferate and avoid apoptosis. The focus of this review covers the principles, advantages, drawbacks and summarize criteria that must be met for design of small molecule-drug conjugates (SMDCs) to achieve the desired therapeutic potency with minimal toxicity. SMDCs are composed of a targeting ligand, a releasable bridge, a spacer, and a therapeutic payload. We summarize the criteria for the effective design that influences the selection of tumor specific receptor and optimum elements in the design of SMDCs. We also discuss the criteria for selecting the optimal therapeutic drug payload, spacer and linker. The linker chemistries and cleavage strategies are also discussed. Finally, we review the folate receptor targeting SMDCs that are in preclinical development and in clinical trials.
Collapse
Affiliation(s)
- Abhilash Rana
- Amity Institute of Biotechnology, Amity University, Sector125, Noida, Uttar Pradesh, India.
| | - Seema Bhatnagar
- Amity Institute of Biotechnology, Amity University, Sector125, Noida, Uttar Pradesh, India.
| |
Collapse
|
4
|
Banerjee K, Núñez FJ, Haase S, McClellan BL, Faisal SM, Carney SV, Yu J, Alghamri MS, Asad AS, Candia AJN, Varela ML, Candolfi M, Lowenstein PR, Castro MG. Current Approaches for Glioma Gene Therapy and Virotherapy. Front Mol Neurosci 2021; 14:621831. [PMID: 33790740 PMCID: PMC8006286 DOI: 10.3389/fnmol.2021.621831] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 02/16/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive primary brain tumor in the adult population and it carries a dismal prognosis. Inefficient drug delivery across the blood brain barrier (BBB), an immunosuppressive tumor microenvironment (TME) and development of drug resistance are key barriers to successful glioma treatment. Since gliomas occur through sequential acquisition of genetic alterations, gene therapy, which enables to modification of the genetic make-up of target cells, appears to be a promising approach to overcome the obstacles encountered by current therapeutic strategies. Gene therapy is a rapidly evolving field with the ultimate goal of achieving specific delivery of therapeutic molecules using either viral or non-viral delivery vehicles. Gene therapy can also be used to enhance immune responses to tumor antigens, reprogram the TME aiming at blocking glioma-mediated immunosuppression and normalize angiogenesis. Nano-particles-mediated gene therapy is currently being developed to overcome the BBB for glioma treatment. Another approach to enhance the anti-glioma efficacy is the implementation of viro-immunotherapy using oncolytic viruses, which are immunogenic. Oncolytic viruses kill tumor cells due to cancer cell-specific viral replication, and can also initiate an anti-tumor immunity. However, concerns still remain related to off target effects, and therapeutic and transduction efficiency. In this review, we describe the rationale and strategies as well as advantages and disadvantages of current gene therapy approaches against gliomas in clinical and preclinical studies. This includes different delivery systems comprising of viral, and non-viral delivery platforms along with suicide/prodrug, oncolytic, cytokine, and tumor suppressor-mediated gene therapy approaches. In addition, advances in glioma treatment through BBB-disruptive gene therapy and anti-EGFRvIII/VEGFR gene therapy are also discussed. Finally, we discuss the results of gene therapy-mediated human clinical trials for gliomas. In summary, we highlight the progress, prospects and remaining challenges of gene therapies aiming at broadening our understanding and highlighting the therapeutic arsenal for GBM.
Collapse
Affiliation(s)
- Kaushik Banerjee
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Felipe J. Núñez
- Laboratory of Molecular and Cellular Therapy, Fundación Instituto Leloir, Buenos Aires, Argentina
| | - Santiago Haase
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Brandon L. McClellan
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Immunology Graduate Program, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Syed M. Faisal
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Stephen V. Carney
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Cancer Biology Graduate Program, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Jin Yu
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Mahmoud S. Alghamri
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Antonela S. Asad
- Departamento de Biología e Histología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alejandro J. Nicola Candia
- Departamento de Biología e Histología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Maria Luisa Varela
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Marianela Candolfi
- Departamento de Biología e Histología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Pedro R. Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Maria G. Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
5
|
Targeted Delivery of Nanoparticulate Cytochrome C into Glioma Cells Through the Proton-Coupled Folate Transporter. Biomolecules 2019; 9:biom9040154. [PMID: 31003476 PMCID: PMC6523331 DOI: 10.3390/biom9040154] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/11/2019] [Accepted: 04/15/2019] [Indexed: 01/14/2023] Open
Abstract
In this study, we identified the proton-coupled folate transporter (PCFT) as a route for targeted delivery of drugs to some gliomas. Using the techniques of confocal imaging, quantitative reverse transcription-polymerase chain reaction (qRT-PCR), and small interfering (siRNA) knockdown against the PCFT, we demonstrated that Gl261 and A172 glioma cells, but not U87 and primary cultured astrocytes, express the PCFT, which provides selective internalization of folic acid (FA)-conjugated cytochrome c-containing nanoparticles (FA-Cyt c NPs), followed by cell death. The FA-Cyt c NPs (100 µg/mL), had no cytotoxic effects in astrocytes but caused death in glioma cells, according to their level of expression of PCFT. Whole-cell patch clamp recording revealed FA-induced membrane currents in FA-Cyt c NPs-sensitive gliomas, that were reduced by siRNA PCFT knockdown in a similar manner as by application of FA-Cyt c NPs, indicating that the PCFT is a route for internalization of FA-conjugated NPs in these glioma cells. Analysis of human glioblastoma specimens revealed that at least 25% of glioblastomas express elevated level of either PCFT or folate receptor (FOLR1). We conclude that the PCFT provides a mechanism for targeted delivery of drugs to some gliomas as a starting point for the development of efficient methods for treating gliomas with high expression of PCFT and/or FOLR1.
Collapse
|
6
|
Chen S, Le T, Harley BAC, Imoukhuede PI. Characterizing Glioblastoma Heterogeneity via Single-Cell Receptor Quantification. Front Bioeng Biotechnol 2018; 6:92. [PMID: 30050899 PMCID: PMC6050407 DOI: 10.3389/fbioe.2018.00092] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 06/21/2018] [Indexed: 01/09/2023] Open
Abstract
Dysregulation of tyrosine kinase receptor (RTK) signaling pathways play important roles in glioblastoma (GBM). However, therapies targeting these signaling pathways have not been successful, partially because of drug resistance. Increasing evidence suggests that tumor heterogeneity, more specifically, GBM-associated stem and endothelial cell heterogeneity, may contribute to drug resistance. In this perspective article, we introduce a high-throughput, quantitative approach to profile plasma membrane RTKs on single cells. First, we review the roles of RTKs in cancer. Then, we discuss the sources of cell heterogeneity in GBM, providing context to the key cells directing resistance to drugs. Finally, we present our provisionally patented qFlow cytometry approach, and report results of a "proof of concept" patient-derived xenograft GBM study.
Collapse
Affiliation(s)
- Si Chen
- Department of Bioengineering, University of Illinois at Urbana–Champaign, Champaign, IL, United States
| | - Thien Le
- Department of Mathematics and Department of Computer Science, University of Illinois at Urbana–Champaign, Champaign, IL, United States
| | - Brendan A. C. Harley
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana–Champaign, Urbana, IL, United States
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana Champaign, Urbana, IL, United States
| | - P. I. Imoukhuede
- Department of Bioengineering, University of Illinois at Urbana–Champaign, Champaign, IL, United States
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana Champaign, Urbana, IL, United States
- Department of Biomedical Engineering, Washington University, St. Louis, MO, United States
| |
Collapse
|
7
|
Recombinant Immunotoxin Therapy of Glioblastoma: Smart Design, Key Findings, and Specific Challenges. BIOMED RESEARCH INTERNATIONAL 2017; 2017:7929286. [PMID: 28752098 PMCID: PMC5511670 DOI: 10.1155/2017/7929286] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 05/29/2017] [Indexed: 12/23/2022]
Abstract
Recombinant immunotoxins (RITs) refer to a group of recombinant protein-based therapeutics, which consists of two components: an antibody variable fragment or a specific ligand that allows RITs to bind specifically to target cells and an engineered toxin fragment that kills the target cells upon internalization. To date, over 1,000 RITs have been generated and significant success has been achieved in the therapy of hematological malignancies. However, the immunogenicity and off-target toxicities of RITs remain as significant barriers for their application to solid tumor therapy. A group of RITs have also been generated for the treatment of glioblastoma multiforme, and some have demonstrated evidence of tumor response and an acceptable profile of toxicity and safety in early clinical trials. Different from other solid tumors, how to efficiently deliver the RITs to intracranial tumors is more critical and needs to be solved urgently. In this article, we first review the design and expression of RITs, then summarize the key findings in the preclinical and clinical development of RIT therapy of glioblastoma multiforme, and lastly discuss the specific issues that still remain to forward RIT therapy to clinical practice.
Collapse
|
8
|
Wang X, Bodman A, Shi C, Guo D, Wang L, Luo J, Hall WA. Tunable Lipidoid-Telodendrimer Hybrid Nanoparticles for Intracellular Protein Delivery in Brain Tumor Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:4185-92. [PMID: 27375237 PMCID: PMC4982832 DOI: 10.1002/smll.201601234] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 05/27/2016] [Indexed: 05/15/2023]
Abstract
A strategy to precisely engineer lipidoid-telodendrimer binary hybrid nanoparticles that offer enhanced cell membrane permeability for therapeutic proteins to reach the intracellular targets is established. The highly controllable biochemical and physical properties of the nanoparticles make them promising for protein-based brain cancer treatment with the assistance of convection-enhanced delivery.
Collapse
Affiliation(s)
- Xu Wang
- Department of Pharmacology, Upstate Cancer Center, State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | - Alexa Bodman
- Department of Neurosurgery, State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | - Changying Shi
- Department of Pharmacology, Upstate Cancer Center, State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | - Dandan Guo
- Department of Pharmacology, Upstate Cancer Center, State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | - Lili Wang
- Department of Pharmacology, Upstate Cancer Center, State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | | | - Walter A. Hall
- Department of Neurosurgery, State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
9
|
Spangler JB, Moraga I, Mendoza JL, Garcia KC. Insights into cytokine-receptor interactions from cytokine engineering. Annu Rev Immunol 2014; 33:139-67. [PMID: 25493332 DOI: 10.1146/annurev-immunol-032713-120211] [Citation(s) in RCA: 192] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cytokines exert a vast array of immunoregulatory actions critical to human biology and disease. However, the desired immunotherapeutic effects of native cytokines are often mitigated by toxicity or lack of efficacy, either of which results from cytokine receptor pleiotropy and/or undesired activation of off-target cells. As our understanding of the structural principles of cytokine-receptor interactions has advanced, mechanism-based manipulation of cytokine signaling through protein engineering has become an increasingly feasible and powerful approach. Modified cytokines, both agonists and antagonists, have been engineered with narrowed target cell specificities, and they have also yielded important mechanistic insights into cytokine biology and signaling. Here we review the theory and practice of cytokine engineering and rationalize the mechanisms of several engineered cytokines in the context of structure. We discuss specific examples of how structure-based cytokine engineering has opened new opportunities for cytokines as drugs, with a focus on the immunotherapeutic cytokines interferon, interleukin-2, and interleukin-4.
Collapse
Affiliation(s)
- Jamie B Spangler
- Howard Hughes Medical Institute, Department of Molecular and Cellular Physiology, Department of Structural Biology, Stanford University School of Medicine, Stanford, California 94305; , , ,
| | | | | | | |
Collapse
|
10
|
Akhtar MJ, Ahamed M, Alhadlaq HA, Alrokayan SA, Kumar S. Targeted anticancer therapy: Overexpressed receptors and nanotechnology. Clin Chim Acta 2014; 436:78-92. [DOI: 10.1016/j.cca.2014.05.004] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 05/07/2014] [Accepted: 05/10/2014] [Indexed: 01/05/2023]
|