1
|
Bayoumi M, Youshia J, Arafa MG, Nasr M, Sammour OA. Nanocarriers for the treatment of glioblastoma multiforme: A succinct review of conventional and repositioned drugs in the last decade. Arch Pharm (Weinheim) 2024; 357:e2400343. [PMID: 39074966 DOI: 10.1002/ardp.202400343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/31/2024]
Abstract
Glioblastoma multiforme is a very combative and threatening type of cancer. The standard course of treatment involves excising the tumor surgically, then administering chemotherapy and radiation therapy. Because of the presence of the blood-brain barrier and the unique characteristics of the tumor microenvironment, chemotherapy is extremely difficult and has a high incidence of relapse. With their capacity to precisely target and transport therapeutic medications to the tumor while overcoming the challenges provided by invasive and infiltrative gliomas, nanocarriers offer a potentially beneficial treatment option for gliomas. Drug repositioning or, in other words, finding novel therapeutic uses for medications that have received approval for previous uses has also recently emerged to provide alternative treatments for many diseases, with glioblastoma being among them. In this article, our goal is to shed light on the pathogenesis of glioma and summarize the proposed treatment approaches in the last decade, highlighting how combining repositioned drugs and nanocarriers technology can reduce drug resistance and improve therapeutic efficacy in primary glioma.
Collapse
Affiliation(s)
- Mahitab Bayoumi
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| | - John Youshia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mona G Arafa
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
- Chemotherapeutic Unit, Mansoura University Hospitals, Mansoura, Egypt
- Nanotechnology Research Center, The British University in Egypt, Cairo, Egypt
| | - Maha Nasr
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Omaima A Sammour
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
2
|
Serra R, Smith SJ, Rowlinson J, Gorelick N, Moloney C, McCrorie P, Veal GJ, Berry P, Chalmers AJ, Suk I, Shakesheff KM, Alexander C, Grundy RG, Brem H, Tyler BM, Rahman R. Neurosurgical application of olaparib from a thermo-responsive paste potentiates DNA damage to prolong survival in malignant glioma. Br J Cancer 2024:10.1038/s41416-024-02878-2. [PMID: 39433869 DOI: 10.1038/s41416-024-02878-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND There is increased pan-cancer specific interest in repurposing the poly adenosine diphosphate-ribose polymerase-1 (PARP-1) inhibitor, olaparib, for newly diagnosed or recurrent isocitrate dehydrogenase wild type glioblastoma. We explore whether intra-cavity delivery of olaparib confers a survival benefit in a pre-clinical high-grade glioma model. METHODS Primary tumor RNA sequencing data was used to determine PARP-1 as a target in the glioblastoma infiltrative margin. We assessed radiosensitization conferred by olaparib alone and concomitant to genotoxic insults in vitro using clonal growth assays, cell cycle analysis and immunocytochemistry, and in vivo upon post-surgical delivery from a temperature-sensitive polymeric paste. RESULTS RNA-sequencing confirmed PARP-1 as a viable therapy target in glioblastoma infiltrative disease. Acute exposure of glioma cells to olaparib impaired proliferation and induced late-stage apoptosis associated with DNA damage in vitro, potentiated by radiation. Using high-grade glioma orthotopic allografts, a long-term overall survival benefit was observed upon interstitial olaparib delivery concomitant with radiotherapy, compared to systemic olaparib and standard glioblastoma treatment. Combined delivery of olaparib with either temozolomide or etoposide increased long-term survival, suggestive of olaparib functioning as DNA damage sensitizer. CONCLUSIONS Collectively, our data support a rationale for localized olaparib delivery concomitant with the current clinical regimen for malignant glioma treatment.
Collapse
Affiliation(s)
- Riccardo Serra
- Department of Neurosurgery, Johns Hopkins University, Baltimore, USA
| | - Stuart J Smith
- Children's Brain Tumour Research Centre, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Jonathan Rowlinson
- Children's Brain Tumour Research Centre, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Noah Gorelick
- Department of Neurosurgery, Johns Hopkins University, Baltimore, USA
| | - Cara Moloney
- Children's Brain Tumour Research Centre, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Phoebe McCrorie
- Children's Brain Tumour Research Centre, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Gareth J Veal
- Newcastle University Centre for Cancer, Newcastle University, Newcastle, UK
| | - Philip Berry
- Newcastle University Centre for Cancer, Newcastle University, Newcastle, UK
| | | | - Ian Suk
- Department of Neurosurgery, Johns Hopkins University, Baltimore, USA
| | | | | | - Richard G Grundy
- Children's Brain Tumour Research Centre, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Henry Brem
- Department of Neurosurgery, Johns Hopkins University, Baltimore, USA
- Departments of Biomedical Engineering, Oncology and Ophthalmology, Johns Hopkins University, Baltimore, USA
| | - Betty M Tyler
- Department of Neurosurgery, Johns Hopkins University, Baltimore, USA.
| | - Ruman Rahman
- Children's Brain Tumour Research Centre, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK.
| |
Collapse
|
3
|
Jones AB, Tuy K, Hawkins CC, Quinn CH, Saad J, Gary SE, Beierle EA, Ding L, Rochlin KM, Lamb LS, Hjelmeland AB. Temozolomide and the PARP Inhibitor Niraparib Enhance Expression of Natural Killer Group 2D Ligand ULBP1 and Gamma-Delta T Cell Cytotoxicity in Glioblastoma. Cancers (Basel) 2024; 16:2852. [PMID: 39199623 PMCID: PMC11352900 DOI: 10.3390/cancers16162852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/18/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
Glioblastoma (GBM) is an immunologically cold tumor, but several immunotherapy-based strategies show promise, including the administration of ex vivo expanded and activated cytotoxic gamma delta T cells. Cytotoxicity is partially mediated through interactions with natural killer group 2D ligands (NKG2DL) on tumor cells. We sought to determine whether the addition of the blood-brain barrier penetrant PARP inhibitor niraparib to the standard of care DNA alkylator temozolomide (TMZ) could upregulate NKG2DL, thereby improving immune cell recognition. Changes in viability were consistent with prior publications as there was a growth inhibitory effect of the combination of TMZ and niraparib. However, decreases in viability did not always correlate with changes in NKG2DL mRNA. ULBP1/Mult-1 mRNA was increased with the combination therapy in comparison to either drug alone in two of the three cell types tested, even though viability was consistently decreased. mRNA expression correlated with protein levels and ULBP1/MULT-1 cell surface protein was significantly increased with TMZ and niraparib treatment in four of the five cell types tested. Gamma delta T cell-mediated cytotoxicity at a 10:1 effector-to-target ratio was significantly increased upon pretreatment of cells derived from a GBM PDX with TMZ and niraparib in comparison to the control or either drug alone. Together, these data demonstrate that the combination of PARP inhibition, DNA alkylation, and gamma delta T cell therapy has the potential for the treatment of GBM.
Collapse
Affiliation(s)
- Amber B. Jones
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.B.J.); (K.T.); (C.C.H.); (J.S.); (S.E.G.)
| | - Kaysaw Tuy
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.B.J.); (K.T.); (C.C.H.); (J.S.); (S.E.G.)
| | - Cyntanna C. Hawkins
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.B.J.); (K.T.); (C.C.H.); (J.S.); (S.E.G.)
| | - Colin H. Quinn
- Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Joelle Saad
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.B.J.); (K.T.); (C.C.H.); (J.S.); (S.E.G.)
| | - Sam E. Gary
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.B.J.); (K.T.); (C.C.H.); (J.S.); (S.E.G.)
- Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Elizabeth A. Beierle
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Lei Ding
- In8Bio, Inc., New York, NY 10118, USA; (L.D.); (K.M.R.); (L.S.L.)
| | - Kate M. Rochlin
- In8Bio, Inc., New York, NY 10118, USA; (L.D.); (K.M.R.); (L.S.L.)
| | - Lawrence S. Lamb
- In8Bio, Inc., New York, NY 10118, USA; (L.D.); (K.M.R.); (L.S.L.)
| | - Anita B. Hjelmeland
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.B.J.); (K.T.); (C.C.H.); (J.S.); (S.E.G.)
| |
Collapse
|
4
|
Obrador E, Moreno-Murciano P, Oriol-Caballo M, López-Blanch R, Pineda B, Gutiérrez-Arroyo JL, Loras A, Gonzalez-Bonet LG, Martinez-Cadenas C, Estrela JM, Marqués-Torrejón MÁ. Glioblastoma Therapy: Past, Present and Future. Int J Mol Sci 2024; 25:2529. [PMID: 38473776 PMCID: PMC10931797 DOI: 10.3390/ijms25052529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/10/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Glioblastoma (GB) stands out as the most prevalent and lethal form of brain cancer. Although great efforts have been made by clinicians and researchers, no significant improvement in survival has been achieved since the Stupp protocol became the standard of care (SOC) in 2005. Despite multimodality treatments, recurrence is almost universal with survival rates under 2 years after diagnosis. Here, we discuss the recent progress in our understanding of GB pathophysiology, in particular, the importance of glioma stem cells (GSCs), the tumor microenvironment conditions, and epigenetic mechanisms involved in GB growth, aggressiveness and recurrence. The discussion on therapeutic strategies first covers the SOC treatment and targeted therapies that have been shown to interfere with different signaling pathways (pRB/CDK4/RB1/P16ink4, TP53/MDM2/P14arf, PI3k/Akt-PTEN, RAS/RAF/MEK, PARP) involved in GB tumorigenesis, pathophysiology, and treatment resistance acquisition. Below, we analyze several immunotherapeutic approaches (i.e., checkpoint inhibitors, vaccines, CAR-modified NK or T cells, oncolytic virotherapy) that have been used in an attempt to enhance the immune response against GB, and thereby avoid recidivism or increase survival of GB patients. Finally, we present treatment attempts made using nanotherapies (nanometric structures having active anti-GB agents such as antibodies, chemotherapeutic/anti-angiogenic drugs or sensitizers, radionuclides, and molecules that target GB cellular receptors or open the blood-brain barrier) and non-ionizing energies (laser interstitial thermal therapy, high/low intensity focused ultrasounds, photodynamic/sonodynamic therapies and electroporation). The aim of this review is to discuss the advances and limitations of the current therapies and to present novel approaches that are under development or following clinical trials.
Collapse
Affiliation(s)
- Elena Obrador
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Paz Moreno-Murciano
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
| | - María Oriol-Caballo
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Rafael López-Blanch
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Begoña Pineda
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Julia Lara Gutiérrez-Arroyo
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - Alba Loras
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - Luis G. Gonzalez-Bonet
- Department of Neurosurgery, Castellon General University Hospital, 12004 Castellon, Spain;
| | - Conrado Martinez-Cadenas
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - José M. Estrela
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
- Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Burjassot, Spain
| | | |
Collapse
|
5
|
Li XX, Xu JK, Su WJ, Wu HL, Zhao K, Zhang CM, Chen XK, Yang LX. The role of KDM4A-mediated histone methylation on temozolomide resistance in glioma cells through the HUWE1/ROCK2 axis. Kaohsiung J Med Sci 2024; 40:161-174. [PMID: 37873881 DOI: 10.1002/kjm2.12768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/05/2023] [Accepted: 09/20/2023] [Indexed: 10/25/2023] Open
Abstract
Temozolomide (TMZ) resistance presents a significant challenge in the treatment of gliomas. Although lysine demethylase 4A (KDM4A) has been implicated in various cancer-related processes, its role in TMZ resistance remains unclear. This study aims to elucidate the contribution of KDM4A to TMZ resistance in glioma cells and its potential implications for glioma prognosis. We assessed the expression of KDM4A in glioma cells (T98G and U251MG) using qRT-PCR and Western blot assays. To explore the role of KDM4A in TMZ resistance, we transfected siRNA targeting KDM4A into drug-resistant glioma cells. Cell viability was assessed using the CCK-8 assay and the TMZ IC50 value was determined. ChIP assays were conducted to investigate KDM4A, H3K9me3, and H3K36me3 enrichment on the promoters of ROCK2 and HUWE1. Co-immunoprecipitation confirmed the interaction between HUWE1 and ROCK2, and we examined the levels of ROCK2 ubiquitination following MG132 treatment. Notably, T98G cells exhibited greater resistance to TMZ than U251MG cells, and KDM4A displayed high expression in T98G cells. Inhibiting KDM4A resulted in decreased cell viability and a reduction in the TMZ IC50 value. Mechanistically, KDM4A promoted ROCK2 transcription by modulating H3K9me3 levels. Moreover, disruption of the interaction between HUWE1 and ROCK2 led to reduced ROCK2 ubiquitination. Inhibition of HUWE1 or overexpression of ROCK2 counteracted the sensitization effect of si-KDM4A on TMZ responsiveness in T98G cells. Our findings highlight KDM4A's role in enhancing TMZ resistance in glioma cells by modulating ROCK2 and HUWE1 transcription and expression through H3K9me3 and H3K36me3 removal.
Collapse
Affiliation(s)
- Xi-Xi Li
- Department of Neurosurgery, Sun Yat-sen University 1st Affiliated Hospital, Guangzhou, China
| | - Jia-Kun Xu
- Department of Neurosurgery, Sun Yat-sen University 1st Affiliated Hospital, Guangzhou, China
| | - Wei-Jie Su
- Department of Neurosurgery, Sun Yat-sen University 1st Affiliated Hospital, Guangzhou, China
| | - Hong-Lin Wu
- Department of Neurosurgery, Sun Yat-sen University 1st Affiliated Hospital, Guangzhou, China
| | - Kun Zhao
- Department of Neurosurgery, Sun Yat-sen University 1st Affiliated Hospital, Guangzhou, China
| | - Chang-Ming Zhang
- Department of Neurosurgery, Sun Yat-sen University 1st Affiliated Hospital, Guangzhou, China
| | - Xiang-Kun Chen
- Department of Emergency, The First People's Hospital of Suqian, Suqian, China
| | - Li-Xuan Yang
- Department of Neurosurgery, Sun Yat-sen University 1st Affiliated Hospital, Guangzhou, China
| |
Collapse
|
6
|
Kosianova А, Pak O, Bryukhovetskiy I. Regulation of cancer stem cells and immunotherapy of glioblastoma (Review). Biomed Rep 2024; 20:24. [PMID: 38170016 PMCID: PMC10758921 DOI: 10.3892/br.2023.1712] [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: 09/06/2023] [Accepted: 11/24/2023] [Indexed: 01/05/2024] Open
Abstract
Glioblastoma (GB) is one of the most adverse diagnoses in oncology. Complex current treatment results in a median survival of 15 months. Resistance to treatment is associated with the presence of cancer stem cells (CSCs). The present review aimed to analyze the mechanisms of CSC plasticity, showing the particular role of β-catenin in regulating vital functions of CSCs, and to describe the molecular mechanisms of Wnt-independent increase of β-catenin levels, which is influenced by the local microenvironment of CSCs. The present review also analyzed the reasons for the low effectiveness of using medication in the regulation of CSCs, and proposed the development of immunotherapy scenarios with tumor cell vaccines, containing heterogenous cancer cells able of producing a multidirectional antineoplastic immune response. Additionally, the possibility of managing lymphopenia by transplanting hematopoietic stem cells from a healthy sibling and using clofazimine or other repurposed drugs that reduce β-catenin concentration in CSCs was discussed in the present study.
Collapse
Affiliation(s)
- Аleksandra Kosianova
- Medical Center, School of Medicine and Life Science, Far Eastern Federal University, Vladivostok 690091, Russian Federation
| | - Oleg Pak
- Medical Center, School of Medicine and Life Science, Far Eastern Federal University, Vladivostok 690091, Russian Federation
| | - Igor Bryukhovetskiy
- Medical Center, School of Medicine and Life Science, Far Eastern Federal University, Vladivostok 690091, Russian Federation
| |
Collapse
|
7
|
Cecchini M, Zhang JY, Wei W, Sklar J, Lacy J, Zhong M, Kong Y, Zhao H, DiPalermo J, Devine L, Stein SM, Kortmansky J, Johung KL, Bindra RS, LoRusso P, Schalper KA. Quantitative DNA Repair Biomarkers and Immune Profiling for Temozolomide and Olaparib in Metastatic Colorectal Cancer. CANCER RESEARCH COMMUNICATIONS 2023; 3:1132-1139. [PMID: 37387791 PMCID: PMC10305782 DOI: 10.1158/2767-9764.crc-23-0045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 03/28/2023] [Accepted: 05/30/2023] [Indexed: 07/01/2023]
Abstract
Purpose O6-methylguanine DNA methyltransferase (MGMT)-silenced tumors reveal sensitivity to temozolomide (TMZ), which may be enhanced by PARP inhibitors. Approximately 40% of colorectal cancer has MGMT silencing and we aimed to measure antitumoral and immunomodulatory effects from TMZ and olaparib in colorectal cancer. Experimental Design Patients with advanced colorectal cancer were screened for MGMT promoter hypermethylation using methylation-specific PCR of archival tumor. Eligible patients received TMZ 75 mg/m2 days 1-7 with olaparib 150 mg twice daily every 21 days. Pretreatment tumor biopsies were collected for whole-exome sequencing (WES), and multiplex quantitative immunofluorescence (QIF) of MGMT protein expression and immune markers. Results MGMT promoter hypermethylation was detected in 18/51 (35%) patients, 9 received study treatment with no objective responses, 5/9 had stable disease (SD) and 4/9 had progressive disease as best response. Three patients had clinical benefit: carcinoembryonic antigen reduction, radiographic tumor regression, and prolonged SD. MGMT expression by multiplex QIF revealed prominent tumor MGMT protein from 6/9 patients without benefit, while MGMT protein was lower in 3/9 with benefit. Moreover, benefitting patients had higher baseline CD8+ tumor-infiltrating lymphocytes. WES revealed 8/9 patients with MAP kinase variants (7 KRAS and 1 ERBB2). Flow cytometry identified peripheral expansion of effector T cells. Conclusions Our results indicate discordance between MGMT promoter hypermethylation and MGMT protein expression. Antitumor activity seen in patients with low MGMT protein expression, supports MGMT protein as a predictor of alkylator sensitivity. Increased CD8+ TILs and peripheral activated T cells, suggest a role for immunostimulatory combinations. Significance TMZ and PARP inhibitors synergize in vitro and in vivo in tumors with MGMT silencing. Up to 40% of colorectal cancer is MGMT promoter hypermethylated, and we investigated whether TMZ and olaparib are effective in this population. We also measured MGMT by QIF and observed efficacy only in patients with low MGMT, suggesting quantitative MGMT biomarkers more accurately predict benefit to alkylator combinations.
Collapse
Affiliation(s)
- Michael Cecchini
- Department of Internal Medicine (Medical Oncology), Yale University School of Medicine, New Haven, Connecticut
| | - Janie Y. Zhang
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Wei Wei
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut
| | - Jeffrey Sklar
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Jill Lacy
- Department of Internal Medicine (Medical Oncology), Yale University School of Medicine, New Haven, Connecticut
| | - Minghao Zhong
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Yong Kong
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut
| | - Jassim DiPalermo
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Lesley Devine
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Stacey M. Stein
- Department of Internal Medicine (Medical Oncology), Yale University School of Medicine, New Haven, Connecticut
| | - Jeremy Kortmansky
- Department of Internal Medicine (Medical Oncology), Yale University School of Medicine, New Haven, Connecticut
| | - Kimberly L. Johung
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Ranjit S. Bindra
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Patricia LoRusso
- Department of Internal Medicine (Medical Oncology), Yale University School of Medicine, New Haven, Connecticut
| | - Kurt A. Schalper
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
8
|
Moran J, Mylod E, Kane LE, Marion C, Keenan E, Mekhaeil M, Lysaght J, Dev KK, O’Sullivan J, Conroy MJ. Investigating the Effects of Olaparib on the Susceptibility of Glioblastoma Multiforme Tumour Cells to Natural Killer Cell-Mediated Responses. Pharmaceutics 2023; 15:360. [PMID: 36839682 PMCID: PMC9959685 DOI: 10.3390/pharmaceutics15020360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/09/2023] [Accepted: 01/14/2023] [Indexed: 01/24/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common adult primary brain malignancy, with dismal survival rates of ~14.6 months. The current standard-of-care consists of surgical resection and chemoradiotherapy, however the treatment response is limited by factors such as tumour heterogeneity, treatment resistance, the blood-brain barrier, and immunosuppression. Several immunotherapies have undergone clinical development for GBM but demonstrated inadequate efficacy, yet future combinatorial approaches are likely to hold more promise. Olaparib is FDA-approved for BRCA-mutated advanced ovarian and breast cancer, and clinical studies have revealed its utility as a safe and efficacious radio- and chemo-sensitiser in GBM. The ability of Olaparib to enhance natural killer (NK) cell-mediated responses has been reported in prostate, breast, and lung cancer. This study examined its potential combination with NK cell therapies in GBM by firstly investigating the susceptibility of the GBM cell line T98G to NK cells and, secondly, examining whether Olaparib can sensitise T98G cells to NK cell-mediated responses. Here, we characterise the NK receptor ligand profile of T98G cells and demonstrate that Olaparib does not dampen T98G susceptibility to NK cells or elicit immunomodulatory effects on the function of NK cells. This study provides novel insights into the potential combination of Olaparib with NK cell therapies for GBM.
Collapse
Affiliation(s)
- Jennifer Moran
- Cancer Immunology Research Group, Department of Physiology, Trinity College Dublin, D02 R590 Dublin, Ireland
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute and Trinity St. James’s Cancer Institute, St. James’s Hospital, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Eimear Mylod
- Cancer Immunology Research Group, Department of Physiology, Trinity College Dublin, D02 R590 Dublin, Ireland
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute and Trinity St. James’s Cancer Institute, St. James’s Hospital, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Laura E. Kane
- Department of Surgery, Trinity Translational Medicine Institute and Trinity St. James’s Cancer Institute, St. James’s Hospital, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Caroline Marion
- Cancer Immunology Research Group, Department of Physiology, Trinity College Dublin, D02 R590 Dublin, Ireland
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute and Trinity St. James’s Cancer Institute, St. James’s Hospital, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Emily Keenan
- Cancer Immunology Research Group, Department of Physiology, Trinity College Dublin, D02 R590 Dublin, Ireland
| | - Marianna Mekhaeil
- Drug Development Research Group, Department of Physiology, School of Medicine, Trinity College Dublin, D02 R590 Dublin, Ireland
| | - Joanne Lysaght
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute and Trinity St. James’s Cancer Institute, St. James’s Hospital, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Kumlesh K. Dev
- Drug Development Research Group, Department of Physiology, School of Medicine, Trinity College Dublin, D02 R590 Dublin, Ireland
| | - Jacintha O’Sullivan
- Department of Surgery, Trinity Translational Medicine Institute and Trinity St. James’s Cancer Institute, St. James’s Hospital, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Melissa J. Conroy
- Cancer Immunology Research Group, Department of Physiology, Trinity College Dublin, D02 R590 Dublin, Ireland
| |
Collapse
|
9
|
Thakur A, Faujdar C, Sharma R, Sharma S, Malik B, Nepali K, Liou JP. Glioblastoma: Current Status, Emerging Targets, and Recent Advances. J Med Chem 2022; 65:8596-8685. [PMID: 35786935 PMCID: PMC9297300 DOI: 10.1021/acs.jmedchem.1c01946] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
Glioblastoma (GBM) is a highly malignant
brain tumor characterized
by a heterogeneous population of genetically unstable and highly infiltrative
cells that are resistant to chemotherapy. Although substantial efforts
have been invested in the field of anti-GBM drug discovery in the
past decade, success has primarily been confined to the preclinical
level, and clinical studies have often been hampered due to efficacy-,
selectivity-, or physicochemical property-related issues. Thus, expansion
of the list of molecular targets coupled with a pragmatic design of
new small-molecule inhibitors with central nervous system (CNS)-penetrating
ability is required to steer the wheels of anti-GBM drug discovery
endeavors. This Perspective presents various aspects of drug discovery
(challenges in GBM drug discovery and delivery, therapeutic targets,
and agents under clinical investigation). The comprehensively covered
sections include the recent medicinal chemistry campaigns embarked
upon to validate the potential of numerous enzymes/proteins/receptors
as therapeutic targets in GBM.
Collapse
Affiliation(s)
- Amandeep Thakur
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei 11031, Taiwan
| | - Chetna Faujdar
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida 201307, India
| | - Ram Sharma
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei 11031, Taiwan
| | - Sachin Sharma
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei 11031, Taiwan
| | - Basant Malik
- Department of Sterile Product Development, Research and Development-Unit 2, Jubiliant Generics Ltd., Noida 201301, India
| | - Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei 11031, Taiwan
| | - Jing Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei 11031, Taiwan
| |
Collapse
|
10
|
Maksoud S. The DNA Double-Strand Break Repair in Glioma: Molecular Players and Therapeutic Strategies. Mol Neurobiol 2022; 59:5326-5365. [PMID: 35696013 DOI: 10.1007/s12035-022-02915-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 06/05/2022] [Indexed: 12/12/2022]
Abstract
Gliomas are the most frequent type of tumor in the central nervous system, which exhibit properties that make their treatment difficult, such as cellular infiltration, heterogeneity, and the presence of stem-like cells responsible for tumor recurrence. The response of this type of tumor to chemoradiotherapy is poor, possibly due to a higher repair activity of the genetic material, among other causes. The DNA double-strand breaks are an important type of lesion to the genetic material, which have the potential to trigger processes of cell death or cause gene aberrations that could promote tumorigenesis. This review describes how the different cellular elements regulate the formation of DNA double-strand breaks and their repair in gliomas, discussing the therapeutic potential of the induction of this type of lesion and the suppression of its repair as a control mechanism of brain tumorigenesis.
Collapse
Affiliation(s)
- Semer Maksoud
- Experimental Therapeutics and Molecular Imaging Unit, Department of Neurology, Neuro-Oncology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
11
|
Abstract
ABSTRACT Poly(ADP-ribose) polymerase (PARP) inhibitors have transformed the therapeutic landscape for advanced ovarian cancer and expanded treatment options for other tumor types, including breast, pancreas, and prostate cancer. Yet, despite the success of PARP inhibitors in our current therapeutic armamentarium, not all patients benefit because of primary resistance, whereas different acquired resistance mechanisms can lead to disease progression on therapy. In addition, the toxicity profile of PARP inhibitors, primarily myelosuppression, has led to adverse events in a proportion of patients as monotherapy, and has limited the use of PARP inhibitors for certain rational combination strategies, such as chemotherapy and targeted therapy regimens. Currently approved PARP inhibitors are essentially equipotent against PARP1 and PARP2 enzymes. In this review, we describe the development of next-generation PARP1-selective inhibitors that have entered phase I clinical trials. These inhibitors have demonstrated increased PARP1 inhibitory potency and exquisitely high PARP1 selectivity in preclinical studies-features that may lead to improved clinical efficacy and a wider therapeutic window. First-in-human clinical trials seeking to establish the safety, tolerability, and recommended phase II dose, as well as antitumor activity of these novel agents, have commenced. If successful, this next-generation of PARP1-selective agents promises to build on the succeses of current PARP inhibitor treatment paradigms in cancer medicine.
Collapse
|