1
|
Riva M, Wouters R, Nittner D, Ceusters J, Sterpin E, Giovannoni R, Himmelreich U, Gsell W, VAN Ranst M, Coosemans A. Radiation dose-escalation and dose-fractionation modulate the immune microenvironment, cancer stem cells and vasculature in experimental high-grade gliomas. J Neurosurg Sci 2023; 67:55-65. [PMID: 33056947 DOI: 10.23736/s0390-5616.20.05060-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND In the context of high-grade gliomas (HGGs), very little evidence is available concerning the optimal radiotherapy (RT) schedule to be used in radioimmunotherapy combinations. This studied was aimed at shedding new light in this field by analyzing the effects of RT dose escalation and dose fractionation on the tumor microenvironment of experimental HGGs. METHODS Neurospheres (NS) CT-2A HGG-bearing C57BL/6 mice were treated with stereotactic RT. For dose-escalation experiments, mice received 2, 4 or 8 Gy as single administrations. For dose-fractionation experiments, mice received 4 Gy as a single fraction or multiple (1.33x3 Gy) fractions. The impact of the RT schedule on murine survival and tumor immunity was evaluated. Modifications of glioma stem cells (GSCs), tumor vasculature and tumor cell replication were also assessed. RESULTS RT dose-escalation was associated with an improved immune profile, with higher CD8+ T cells and CD8+ T cells/regulatory T cells (Tregs) ratio (P=0.0003 and P=0.0022, respectively) and lower total tumor associated microglia/macrophages (TAMs), M2 TAMs and monocytic myeloid derived suppressor cells (mMDSCs) (P=0.0011, P=0.0024 and P<0.0001, respectively). The progressive increase of RT dosages prolonged survival (P<0.0001) and reduced tumor vasculature (P=0.069), tumor cell proliferation (P<0.0001) and the amount of GSCs (P=0.0132 or lower). Compared to the unfractionated regimen, RT dose-fractionation negatively affected tumor immunity by inducing higher total TAMs, M2 TAMs and mMDSCs (P=0.0051, P=0.0036 and P=0.0436, respectively). Fractionation also induced a shorter survival (P=0.0078), a higher amount of GSCs (P=0.0015 or lower) and a higher degree of tumor cell proliferation (P=0.0003). CONCLUSIONS This study demonstrates that RT dosage and fractionation significantly influence survival, tumor immunity and GSCs in experimental HGGs. These findings should be taken into account when aiming at designing more synergistic and effective radio-immunotherapy combinations.
Collapse
Affiliation(s)
- Matteo Riva
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Catholic University of Leuven, Leuven, Belgium - .,Department of Neurosurgery, UcL Namur, Mont-Godinne University Hospital, Yvoir, Belgium -
| | - Roxanne Wouters
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Catholic University of Leuven, Leuven, Belgium
| | - David Nittner
- Center for the Biology of Disease, Catholic University of Leuven Center for Human Genetics - InfraMouse, VIB, Catholic University of University of Leuven, Leuven, Belgium
| | - Jolien Ceusters
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Catholic University of Leuven, Leuven, Belgium
| | - Edmond Sterpin
- Laboratory of Experimental Radiotherapy, Department of Oncology, Catholic University of Leuven, Leuven, Belgium
| | | | - Uwe Himmelreich
- Biomedical MRI, Department of Imaging and Pathology and Molecular Small Animal Imaging Center (MoSAIC), Catholic University of Leuven, Leuven, Belgium
| | - Willy Gsell
- Biomedical MRI, Department of Imaging and Pathology and Molecular Small Animal Imaging Center (MoSAIC), Catholic University of Leuven, Leuven, Belgium
| | - Marc VAN Ranst
- Laboratory for Clinical and Epidemiological Virology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - An Coosemans
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Catholic University of Leuven, Leuven, Belgium.,Department of Gynecology and Obstetrics, Leuven Cancer Institute, UZ Leuven, Leuven, Belgium
| |
Collapse
|
2
|
Liu Y, An L, Huang R, Xiong J, Yang H, Wang X, Zhang X. Strategies to enhance CAR-T persistence. Biomark Res 2022; 10:86. [PMID: 36419115 PMCID: PMC9685914 DOI: 10.1186/s40364-022-00434-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/09/2022] [Indexed: 11/25/2022] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy has significantly improved the life expectancy for patients with refractory or relapse B cell lymphoma. As for B cell acute lymphoblastic leukemia (B-ALL), although the primary response rate is promising, the high incidence of early relapse has caused modest long-term survival with CAR-T cell alone. One of the main challenges is the limited persistence of CAR-T cells. To further optimize the clinical effects of CAR-T cells, many studies have focused on modifying the CAR structure and regulating CAR-T cell differentiation. In this review, we focus on CAR-T cell persistence and summarize the latest progress and strategies adopted during the in vitro culture stage to optimize CAR-T immunotherapy by improving long-term persistence. Such strategies include choosing a suitable cell source, improving culture conditions, combining CAR-T cells with conventional drugs, and applying genetic manipulations, all of which may improve the survival of patients with hematologic malignancies by reducing the probability of recurrence after CAR-T cell infusion and provide clues for solid tumor CAR-T cell therapy development.
Collapse
Affiliation(s)
- Yue Liu
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, 400037, Chongqing, China
| | - Lingna An
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, 400037, Chongqing, China
| | - Ruihao Huang
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, 400037, Chongqing, China
| | - Jingkang Xiong
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, 400037, Chongqing, China
| | - Haoyu Yang
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, 400037, Chongqing, China
| | - Xiaoqi Wang
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, 400037, Chongqing, China.
| | - Xi Zhang
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, 400037, Chongqing, China. .,Jinfeng Laboratory, 401329, Chongqing, China.
| |
Collapse
|
3
|
Konig S, Strobel H, Grunert M, Lyszkiewicz M, Brühl O, Karpel-Massler G, Ziętara N, La Ferla-Brühl K, Siegelin MD, Debatin KM, Westhoff MA. Unblinding the watchmaker: cancer treatment and drug design in the face of evolutionary pressure. Expert Opin Drug Discov 2022; 17:1081-1094. [PMID: 35997138 DOI: 10.1080/17460441.2022.2114454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Death due to cancer is mostly associated with therapy ineffectiveness, i.e. tumor cells no longer responding to treatment. The underlying dynamics that facilitate this mutational escape from selective pressure are well studied in several other fields and several interesting approaches exist to combat this phenomenon, for example in the context of antibiotic-resistance in bacteria. AREAS COVERED Ninety percent of all cancer-related deaths are associated with treatment failure. Here, we discuss the common treatment modalities and prior attempts to overcome acquired resistance to therapy. The underlying molecular mechanisms are discussed and the implications of emerging resistance in other systems, such as bacteria, are discussed in the context of cancer. EXPERT OPINION Reevaluating emerging therapy resistance in tumors as an evolutionary mechanism to survive in a rapidly and drastically altering fitness landscape leads to novel treatment strategies and distinct requirements for new drugs. Here, we propose a scheme of considerations that need to be applied prior to the discovery of novel therapeutic drugs.
Collapse
Affiliation(s)
- Sophia Konig
- Department of Pediatrics and Adolescent Medicine, Ulm University Hospital, Ulm, Germany
| | - Hannah Strobel
- Department of Pediatrics and Adolescent Medicine, Ulm University Hospital, Ulm, Germany
| | - Michael Grunert
- Department of Nuclear Medicine, German Armed Forces Hospital of Ulm, Ulm, Germany
| | - Marcin Lyszkiewicz
- Department of Pediatrics and Adolescent Medicine, Ulm University Hospital, Ulm, Germany
| | - Oliver Brühl
- Laboratorio Analisi Sicilia, Catania, Lentini, Italy
| | | | - Natalia Ziętara
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH & Co. KG, Germany
| | | | - Markus D Siegelin
- Department of Pathology and Cell Biology, Columbia University Medical Center, Albany, NY, USA
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, Ulm University Hospital, Ulm, Germany
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, Ulm University Hospital, Ulm, Germany
| |
Collapse
|
4
|
He K, Zhang S, Pang J, Yin JC, Mu D, Wang J, Ge H, Ma J, Yang Z, Zheng X, Dong L, Zhang J, Chang P, Li L, Tang S, Bao H, Wu X, Wang X, Shao Y, Yu J, Yuan S. Genomic Profiling Reveals Novel Predictive Biomarkers for Chemo-Radiotherapy Efficacy and Thoracic Toxicity in Non-Small-Cell Lung Cancer. Front Oncol 2022; 12:928605. [PMID: 35912186 PMCID: PMC9329611 DOI: 10.3389/fonc.2022.928605] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 05/23/2022] [Indexed: 11/23/2022] Open
Abstract
Chemo-radiotherapy (CRT) remains the main treatment modality for non-small-cell lung cancer (NSCLC). However, its clinical efficacy is largely limited by individual variations in radio-sensitivity and radiotherapy-associated toxicity. There is an urgent need to identify genetic determinants that can explain patients’ likelihood to develop recurrence and radiotherapy-associated toxicity following CRT. In this study, we performed comprehensive genomic profiling, using a 474-cancer- and radiotherapy-related gene panel, on pretreatment biopsy samples from patients with unresectable stage III NSCLCs who underwent definitive CRT. Patients’ baseline clinical characteristics and genomic features, including tumor genetic, genomic and molecular pathway alterations, as well as single nucleotide polymorphisms (SNPs), were correlated with progression-free survival (PFS), overall survival (OS), and radiotherapy-associated pneumonitis and/or esophagitis development after CRT. A total of 122 patients were enrolled between 2014 and 2019, with 84 (69%) squamous cell carcinomas and 38 (31%) adenocarcinomas. Genetic analysis confirmed the association between the KEAP1-NRF2 pathway gene alterations and unfavorable survival outcome, and revealed alterations in FGFR family genes, MET, PTEN, and NOTCH2 as potential novel and independent risk factors of poor post-CRT survival. Combined analysis of such alterations led to improved stratification of the risk populations. In addition, patients with EGFR activating mutations or any oncogenic driver mutations exhibited improved OS. On the other hand, we also identified genetic markers in relation to radiotherapy-associated thoracic toxicity. SNPs in the DNA repair-associated XRCC5 (rs3835) and XRCC1 (rs25487) were associated with an increased risk of high-grade esophagitis and pneumonitis respectively. MTHFR (rs1801133) and NQO1 (rs1800566) were additional risk alleles related to higher susceptibility to pneumonitis and esophagitis overall. Moreover, through their roles in genome integrity and replicative fidelity, somatic alterations in ZNF217 and POLD1 might also serve as risk predictors of high-grade pneumonitis and esophagitis. Taken together, leveraging targeted next-generating sequencing, we identified a set of novel clinically applicable biomarkers that might enable prediction of survival outcomes and risk of radiotherapy-associated thoracic toxicities. Our findings highlight the value of pre-treatment genetic testing to better inform CRT outcomes and clinical actions in stage III unresectable NSCLCs.
Collapse
Affiliation(s)
- Kewen He
- Department of Radiation Oncology, Shandong University Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Shaotong Zhang
- Department of Ultrasound, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jiaohui Pang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Jiani C. Yin
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Dianbin Mu
- Department of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jun Wang
- Department of Radiation Oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hong Ge
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Jie Ma
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhe Yang
- Department of Radiation Oncology, Shandong Provincial Hospital, Jinan, China
| | - Xiaoli Zheng
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Lihua Dong
- Department of Radiation Oncology & Therapy, Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Jilin, China
| | - Junli Zhang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Pengyu Chang
- Department of Radiation Oncology & Therapy, Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Jilin, China
| | - Li Li
- Department of Radiation Oncology, Shandong University Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Shanshan Tang
- Department of Radiation Oncology, Shandong University Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Hua Bao
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Xue Wu
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Xiaonan Wang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Yang Shao
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
- School of Public Health, Nanjing Medical University, Nanjing, China
- *Correspondence: Shuanghu Yuan, ; Jinming Yu, ; Yang Shao,
| | - Jinming Yu
- Department of Radiation Oncology, Shandong University Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- *Correspondence: Shuanghu Yuan, ; Jinming Yu, ; Yang Shao,
| | - Shuanghu Yuan
- Department of Radiation Oncology, Shandong University Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- *Correspondence: Shuanghu Yuan, ; Jinming Yu, ; Yang Shao,
| |
Collapse
|
5
|
Petroni G, Cantley LC, Santambrogio L, Formenti SC, Galluzzi L. Radiotherapy as a tool to elicit clinically actionable signalling pathways in cancer. Nat Rev Clin Oncol 2022; 19:114-131. [PMID: 34819622 PMCID: PMC9004227 DOI: 10.1038/s41571-021-00579-w] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2021] [Indexed: 02/03/2023]
Abstract
A variety of targeted anticancer agents have been successfully introduced into clinical practice, largely reflecting their ability to inhibit specific molecular alterations that are required for disease progression. However, not all malignant cells rely on such alterations to survive, proliferate, disseminate and/or evade anticancer immunity, implying that many tumours are intrinsically resistant to targeted therapies. Radiotherapy is well known for its ability to activate cytotoxic signalling pathways that ultimately promote the death of cancer cells, as well as numerous cytoprotective mechanisms that are elicited by cellular damage. Importantly, many cytoprotective mechanisms elicited by radiotherapy can be abrogated by targeted anticancer agents, suggesting that radiotherapy could be harnessed to enhance the clinical efficacy of these drugs. In this Review, we discuss preclinical and clinical data that introduce radiotherapy as a tool to elicit or amplify clinically actionable signalling pathways in patients with cancer.
Collapse
Affiliation(s)
- Giulia Petroni
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Lewis C Cantley
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Laura Santambrogio
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
| | - Silvia C Formenti
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| |
Collapse
|
6
|
Westhoff MA, Schuler-Ortoli M, Zerrinius D, Hadzalic A, Schuster A, Strobel H, Scheuerle A, Wong T, Wirtz CR, Debatin KM, Peraud A. Bcl-XL but Not Bcl-2 Is a Potential Target in Medulloblastoma Therapy. Pharmaceuticals (Basel) 2022; 15:ph15010091. [PMID: 35056150 PMCID: PMC8779796 DOI: 10.3390/ph15010091] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/29/2021] [Accepted: 01/05/2022] [Indexed: 01/26/2023] Open
Abstract
Medulloblastoma (MB) is the most common solid tumour in children and, despite current treatment with a rather aggressive combination therapy, accounts for 10% of all deaths associated with paediatric cancer. Breaking the tumour cells’ intrinsic resistance to therapy-induced cell death should lead to less aggressive and more effective treatment options. In other tumour entities, this has been achieved by modulating the balance between the various pro- and anti-apoptotic members of the Bcl-2 family with small molecule inhibitors. To evaluate the therapeutic benefits of ABT-199 (Venetoclax), a Bcl-2 inhibitor, and ABT-263 (Navitoclax), a dual Bcl-XL/Bcl-2 inhibitor, increasingly more relevant model systems were investigated. Starting from established MB cell lines, progressing to primary patient-derived material and finally an experimental tumour system imbedded in an organic environment were chosen. Assessment of the metabolic activity (a surrogate readout for population viability), the induction of DNA fragmentation (apoptosis) and changes in cell number (the combined effect of alterations in proliferation and cell death induction) revealed that ABT-263, but not ABT-199, is a promising candidate for combination therapy, synergizing with cell death-inducing stimuli. Interestingly, in the experimental tumour setting, the sensitizing effect of ABT-263 seems to be predominantly mediated via an anti-proliferative and not a pro-apoptotic effect, opening a future line of investigation. Our data show that modulation of specific members of the Bcl-2 family might be a promising therapeutic addition for the treatment of MB.
Collapse
Affiliation(s)
- Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, Ulm University Hospital, 89075 Ulm, Germany; (A.S.); (H.S.); (T.W.); (K.-M.D.)
- Correspondence: (M.-A.W.); (A.P.); Tel.: +49-731-500-57495 (M.-A.W.); +49-731-500-55001 (A.P.)
| | - Marie Schuler-Ortoli
- Section Pediatric Neurosurgery, Department of Neurosurgery, Ulm University Hospital, 89081 Ulm, Germany; (M.S.-O.); (D.Z.); (A.H.)
| | - Daniela Zerrinius
- Section Pediatric Neurosurgery, Department of Neurosurgery, Ulm University Hospital, 89081 Ulm, Germany; (M.S.-O.); (D.Z.); (A.H.)
| | - Amina Hadzalic
- Section Pediatric Neurosurgery, Department of Neurosurgery, Ulm University Hospital, 89081 Ulm, Germany; (M.S.-O.); (D.Z.); (A.H.)
| | - Andrea Schuster
- Department of Pediatrics and Adolescent Medicine, Ulm University Hospital, 89075 Ulm, Germany; (A.S.); (H.S.); (T.W.); (K.-M.D.)
| | - Hannah Strobel
- Department of Pediatrics and Adolescent Medicine, Ulm University Hospital, 89075 Ulm, Germany; (A.S.); (H.S.); (T.W.); (K.-M.D.)
| | | | - Tiana Wong
- Department of Pediatrics and Adolescent Medicine, Ulm University Hospital, 89075 Ulm, Germany; (A.S.); (H.S.); (T.W.); (K.-M.D.)
- Section Pediatric Neurosurgery, Department of Neurosurgery, Ulm University Hospital, 89081 Ulm, Germany; (M.S.-O.); (D.Z.); (A.H.)
| | | | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, Ulm University Hospital, 89075 Ulm, Germany; (A.S.); (H.S.); (T.W.); (K.-M.D.)
| | - Aurelia Peraud
- Section Pediatric Neurosurgery, Department of Neurosurgery, Ulm University Hospital, 89081 Ulm, Germany; (M.S.-O.); (D.Z.); (A.H.)
- Correspondence: (M.-A.W.); (A.P.); Tel.: +49-731-500-57495 (M.-A.W.); +49-731-500-55001 (A.P.)
| |
Collapse
|
7
|
Inferring tumor progression in large datasets. PLoS Comput Biol 2020; 16:e1008183. [PMID: 33035204 PMCID: PMC7577444 DOI: 10.1371/journal.pcbi.1008183] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 10/21/2020] [Accepted: 07/22/2020] [Indexed: 12/31/2022] Open
Abstract
Identification of mutations of the genes that give cancer a selective advantage is an important step towards research and clinical objectives. As such, there has been a growing interest in developing methods for identification of driver genes and their temporal order within a single patient (intra-tumor) as well as across a cohort of patients (inter-tumor). In this paper, we develop a probabilistic model for tumor progression, in which the driver genes are clustered into several ordered driver pathways. We develop an efficient inference algorithm that exhibits favorable scalability to the number of genes and samples compared to a previously introduced ILP-based method. Adopting a probabilistic approach also allows principled approaches to model selection and uncertainty quantification. Using a large set of experiments on synthetic datasets, we demonstrate our superior performance compared to the ILP-based method. We also analyze two biological datasets of colorectal and glioblastoma cancers. We emphasize that while the ILP-based method puts many seemingly passenger genes in the driver pathways, our algorithm keeps focused on truly driver genes and outputs more accurate models for cancer progression. Cancer is a disease caused by the accumulation of somatic mutations in the genome. This process is mainly driven by mutations in certain genes that give the harboring cells some selective advantage. The rather few driver genes are usually masked amongst an abundance of so-called passenger mutations. Identification of the driver genes and the temporal order in which the mutations occur is of great importance towards research and clinical objectives. In this paper, we introduce a probabilistic model for cancer progression and devise an efficient inference algorithm to train the model. We show that our method scales favorably to large datasets and provides superior performance compared to an ILP-based counterpart on a wide set of synthetic data simulations. Our Bayesian approach also allows for systematic model selection and confidence quantification procedures in contrast to the previous non-probabilistic progression models. We also study two large datasets on colorectal and glioblastoma cancers and validate our inferred model in comparison to the ILP-based method.
Collapse
|
8
|
The limitations of targeting MEK signalling in Glioblastoma therapy. Sci Rep 2020; 10:7401. [PMID: 32366879 PMCID: PMC7198577 DOI: 10.1038/s41598-020-64289-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 04/15/2020] [Indexed: 01/16/2023] Open
Abstract
Glioblastoma (GB) is a highly aggressive, difficult to treat brain tumour. Successful treatment, consisting of maximal safe tumour de-bulking, followed by radiotherapy and treatment with the alkylating agent Temozolomide (TMZ), can extend patient survival to approximately 15 months. Combination treatments based on the inhibition of the PI3K pathway, which is the most frequently activated signalling cascade in GB, have so far only shown limited therapeutic success. Here, we use the clinically approved MEK inhibitor Trametinib to investigate its potential use in managing GB. Trametinib has a strong anti-proliferative effect on established GB cell lines, stem cell-like cells and their differentiated progeny and while it does not enhance anti-proliferative and cell death-inducing properties of the standard treatment, i.e. exposure to radiation or TMZ, neither does MEK inhibition block their effectiveness. However, upon MEK inhibition some cell populations appear to favour cell-substrate interactions in a sprouting assay and become more invasive in the Chorioallantoic Membrane assay, which assesses cell penetration into an organic membrane. While this increased invasion can be modulated by additional inhibition of the PI3K signalling cascade, there is no apparent benefit of blocking MEK compared to targeting PI3K.
Collapse
|
9
|
Westhoff MA, Baisch T, Herbener VJ, Karpel-Massler G, Debatin KM, Strobel H. Comment in Response to "Temozolomide in Glioblastoma Therapy: Role of Apoptosis, Senescence and Autophagy etc. by B. Kaina". Biomedicines 2020; 8:biomedicines8040093. [PMID: 32326020 PMCID: PMC7235879 DOI: 10.3390/biomedicines8040093] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 04/17/2020] [Indexed: 12/17/2022] Open
Abstract
It is with great pleasure that we acknowledge the fact that our review on Temozolomide (TMZ) has initiated a discussion [1-3]. [...].
Collapse
Affiliation(s)
- Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (T.B.); (V.J.H.); (K.-M.D.); (H.S.)
- Correspondence:
| | - Tim Baisch
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (T.B.); (V.J.H.); (K.-M.D.); (H.S.)
| | - Verena J. Herbener
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (T.B.); (V.J.H.); (K.-M.D.); (H.S.)
| | - Georg Karpel-Massler
- Department of Neurosurgery, University Medical Center Ulm, D-89081 Ulm, Germany;
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (T.B.); (V.J.H.); (K.-M.D.); (H.S.)
| | - Hannah Strobel
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (T.B.); (V.J.H.); (K.-M.D.); (H.S.)
| |
Collapse
|
10
|
Altine B, Gai Y, Han N, Jiang Y, Ji H, Fang H, Niyonkuru A, Bakari KH, Rajab Arnous MM, Liu Q, Zhang Y, Lan X. Preclinical Evaluation of a Fluorine-18 ( 18F)-Labeled Phosphatidylinositol 3-Kinase Inhibitor for Breast Cancer Imaging. Mol Pharm 2019; 16:4563-4571. [PMID: 31553879 DOI: 10.1021/acs.molpharmaceut.9b00690] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Breast cancer is one of the commonest malignancies in women, especially in middle-aged and elderly women. Abnormal activation of the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKt/mTOR) pathway has been found to be involved in breast cancer proliferation. Pictilisib (GDC-0941) is a potent inhibitor of PI3K with high affinity and is undergoing phase 2 clinical trials. In this study, we aimed to develop a noninvasive PI3K radiotracer to help determine the mechanism of the PI3K/AKt/mTOR pathway to aid in diagnosis. We designed a new 18F-radiolabeled radiotracer based on the structure of pictilisib, to evaluate noninvasively abnormal activation of the PI3K/AKT/mTOR pathway. To increase the water solubility, and to decrease hepatobiliary and gastrointestinal uptake of the tracer, pictilisib was modified with triethylene glycol di(p-toluenesulfonate) (TsO-PEG3-OTs) to obtain TsO-PEG3-GDC-0941 as the precursor for 18F labeling. A nonradiolabeled reference compound [19F]-PEG3-GDC-0941 was also prepared. Breast cancer cell lines, MCF-7 and MDA-MB-231, were used as high- and low-expression PI3K models, respectively. PET imaging and ex vivo biodistribution assays of [18F]-PEG3-GDC-0941 in MCF-7 and MDA-MB-231 xenografts were also performed, and the results were compared. The precursor compound and reference standard compound were successfully synthesized and identified using NMR and mass spectroscopy. The 18F radiolabeling was achieved with a high yield (61 ± 1%) at a high molar activity (2100 ± 100 MBq/mg). MicroPET images and biodistribution studies showed a higher uptake of the radiotracer in MCF-7 tumors than in MDA-MB-231 tumors (7.56 ± 1.01%ID/g vs 4.07 ± 0.68%ID/g, 1 h postinjection). Additionally, the MCF-7 tumor uptake was significantly decreased when a blocking dose of GDC-0941 was coinjected, indicating high specificity. The liver was found to be the major excretory organ with 5.82 ± 0.88%ID/g at 30 min postinjection for MCF-7 mice. This radiotracer holds great potential for patient screening, diagnosis, and therapy prediction of PI3K-related diseases.
Collapse
Affiliation(s)
- Bouhari Altine
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China.,Hubei Province Key Laboratory of Molecular Imaging , Wuhan 430022 , China
| | - Yongkang Gai
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China.,Hubei Province Key Laboratory of Molecular Imaging , Wuhan 430022 , China
| | - Na Han
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China.,Hubei Province Key Laboratory of Molecular Imaging , Wuhan 430022 , China
| | - Yaqun Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China.,Hubei Province Key Laboratory of Molecular Imaging , Wuhan 430022 , China
| | - Hao Ji
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China.,Hubei Province Key Laboratory of Molecular Imaging , Wuhan 430022 , China
| | - Hanyi Fang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China.,Hubei Province Key Laboratory of Molecular Imaging , Wuhan 430022 , China
| | - Alexandre Niyonkuru
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China.,Hubei Province Key Laboratory of Molecular Imaging , Wuhan 430022 , China
| | - Khamis Hassan Bakari
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China.,Hubei Province Key Laboratory of Molecular Imaging , Wuhan 430022 , China
| | - Maher Mohamad Rajab Arnous
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China.,Hubei Province Key Laboratory of Molecular Imaging , Wuhan 430022 , China
| | - Qingyao Liu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China.,Hubei Province Key Laboratory of Molecular Imaging , Wuhan 430022 , China
| | - Yongxue Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China.,Hubei Province Key Laboratory of Molecular Imaging , Wuhan 430022 , China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China.,Hubei Province Key Laboratory of Molecular Imaging , Wuhan 430022 , China
| |
Collapse
|
11
|
Strobel H, Baisch T, Fitzel R, Schilberg K, Siegelin MD, Karpel-Massler G, Debatin KM, Westhoff MA. Temozolomide and Other Alkylating Agents in Glioblastoma Therapy. Biomedicines 2019; 7:biomedicines7030069. [PMID: 31505812 PMCID: PMC6783999 DOI: 10.3390/biomedicines7030069] [Citation(s) in RCA: 160] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 09/02/2019] [Indexed: 12/31/2022] Open
Abstract
The alkylating agent temozolomide (TMZ) together with maximal safe bulk resection and focal radiotherapy comprises the standard treatment for glioblastoma (GB), a particularly aggressive and lethal primary brain tumor. GB affects 3.2 in 100,000 people who have an average survival time of around 14 months after presentation. Several key aspects make GB a difficult to treat disease, primarily including the high resistance of tumor cells to cell death-inducing substances or radiation and the combination of the highly invasive nature of the malignancy, i.e., treatment must affect the whole brain, and the protection from drugs of the tumor bulk—or at least of the invading cells—by the blood brain barrier (BBB). TMZ crosses the BBB, but—unlike classic chemotherapeutics—does not induce DNA damage or misalignment of segregating chromosomes directly. It has been described as a DNA alkylating agent, which leads to base mismatches that initiate futile DNA repair cycles; eventually, DNA strand breaks, which in turn induces cell death. However, while much is assumed about the function of TMZ and its mode of action, primary data are actually scarce and often contradictory. To improve GB treatment further, we need to fully understand what TMZ does to the tumor cells and their microenvironment. This is of particular importance, as novel therapeutic approaches are almost always clinically assessed in the presence of standard treatment, i.e., in the presence of TMZ. Therefore, potential pharmacological interactions between TMZ and novel drugs might occur with unforeseeable consequences.
Collapse
Affiliation(s)
- Hannah Strobel
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany.
| | - Tim Baisch
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany.
| | - Rahel Fitzel
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany.
| | | | - Markus D Siegelin
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA.
| | - Georg Karpel-Massler
- Department of Neurosurgery, University Medical Center Ulm, D-89081 Ulm, Germany.
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany.
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany.
| |
Collapse
|