1
|
Cosgrove PA, Bild AH, Dellinger TH, Badie B, Portnow J, Nath A. Single-Cell Transcriptomics Sheds Light on Tumor Evolution: Perspectives from City of Hope's Clinical Trial Teams. J Clin Med 2024; 13:7507. [PMID: 39768430 PMCID: PMC11677125 DOI: 10.3390/jcm13247507] [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: 10/23/2024] [Revised: 11/28/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
Tumor heterogeneity is a significant factor influencing cancer treatment effectiveness and can arise from genetic, epigenetic, and phenotypic variations among cancer cells. Understanding how tumor heterogeneity impacts tumor evolution and therapy response can lead to more effective treatments and improved patient outcomes. Traditional bulk genomic approaches fail to provide insights into cellular-level events, whereas single-cell RNA sequencing (scRNA-seq) offers transcriptomic analysis at the individual cell level, advancing our understanding of tumor growth, progression, and drug response. However, implementing single-cell approaches in clinical trials involves challenges, such as obtaining high-quality cells, technical variability, and the need for complex computational analysis. Effective implementation of single-cell genomics in clinical trials requires a collaborative "Team Medicine" approach, leveraging shared resources, expertise, and workflows. Here, we describe key technical considerations in implementing the collection of research biopsies and lessons learned from integrating scRNA-seq into City of Hope's clinical trial design, highlighting collaborative efforts between computational and clinical teams across breast, brain, and ovarian cancer studies to understand the composition, phenotypic state, and underlying resistance mechanisms within the tumor microenvironment.
Collapse
Affiliation(s)
- Patrick A. Cosgrove
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA; (P.A.C.)
| | - Andrea H. Bild
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA; (P.A.C.)
| | - Thanh H. Dellinger
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Behnam Badie
- Division of Neurosurgery, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Jana Portnow
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA; (P.A.C.)
| | - Aritro Nath
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA; (P.A.C.)
| |
Collapse
|
2
|
Inocencio JF, Mitrasinovic S, Asad M, Parney IF, Zang X, Himes BT. Immune checkpoint pathways in glioblastoma: a diverse and evolving landscape. Front Immunol 2024; 15:1424396. [PMID: 39346924 PMCID: PMC11427296 DOI: 10.3389/fimmu.2024.1424396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 08/27/2024] [Indexed: 10/01/2024] Open
Abstract
Immune checkpoint (IC) inhibition in glioblastoma (GBM) has not shown promising results in the last decade compared to other solid tumors. Several factors contributing to the lack of immunotherapy response include the profound immunosuppressive nature of GBM, highly redundant signaling pathways underlying immune checkpoints, and the negative immunogenic impact of current standard of care on the tumor microenvironment. In this review, we will discuss various ICs in the context of GBM, their interplay with the tumor immune microenvironment, relevant pre-clinical and clinical studies, and the impact of current treatment modalities on GBM IC blockade therapy. Understanding the molecular mechanisms that drive ICs, and how they contribute to an immunosuppressive tumor microenvironment is critical in advancing IC inhibition therapy in GBM. Furthermore, revisiting current treatment modalities and their impact on the immune landscape is instrumental in designing future combinatorial therapies that may overcome treatment resistance.
Collapse
Affiliation(s)
- Julio F Inocencio
- Department of Neurological Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, United States
| | - Stefan Mitrasinovic
- Department of Neurological Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, United States
| | - Mohammad Asad
- Department of Neurological Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, United States
| | - Ian F Parney
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | - Xingxing Zang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
- Department of Oncology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Benjamin T Himes
- Department of Neurological Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, United States
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
3
|
Sanchez-Aceves LM, Pérez-Alvarez I, Onofre-Camarena DB, Gutiérrez-Noya VM, Rosales-Pérez KE, Orozco-Hernández JM, Hernández-Navarro MD, Flores HI, Gómez-Olivan LM. Prolonged exposure to the synthetic glucocorticoid dexamethasone induces brain damage via oxidative stress and apoptotic response in adult Daniorerio. CHEMOSPHERE 2024; 364:143012. [PMID: 39103101 DOI: 10.1016/j.chemosphere.2024.143012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/11/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024]
Abstract
Due to its extensive use as a painkiller, anti-inflammatory, and immune modulatory agent, as well as its effectiveness in treating severe COVID-19, dexamethasone, a synthetic glucocorticoid, has gained attention not only for its impact on public health but also for its environmental implications. Various studies have reported its presence in aquatic environments, including urban waters, surface samples, sediments, drinking water, and wastewater effluents. However, limited information is available regarding its toxic effects on nontarget aquatic organisms. Therefore, this study aimed to investigate the mechanism of toxicity underlying dexamethasone-induced brain damage in the bioindicator Danio rerio following long-term exposure. Adult zebrafish were treated with environmentally relevant concentrations of dexamethasone (20, 40, and 60 ng L-1) for 28 days. To elucidate the possible mechanisms involved in the toxicity of the pharmaceutical compound, we conducted a behavioral test battery (Novel Tank and Light and Dark tests), oxidative stress biomarkers, acetylcholinesterase enzyme activity quantification, histopathological analysis, and gene expression analysis using qRT-PCR (p53, bcl-2, bax, caspase-3, nrf1, and nrf2).The results revealed that the pharmaceutical compound could produce anxiety-like symptoms, increase the oxidative-induced stress response, decrease the activity of acetylcholinesterase enzyme, and cause histopathological alterations, including perineuronal vacuolization, granular and molecular layers deterioration, cell swallowing and intracellular spaces. The expression of genes involved in the apoptotic process (p53, bax, and casp-3) and antioxidant defense (nrf1 and nrf2) was upregulated in response to oxidative damage, while the expression of the anti-apoptotic gene bcl-2 was down-regulated indicating that the environmental presence of dexamethasone may pose a threat to wildlife and human health.
Collapse
Affiliation(s)
- Livier M Sanchez-Aceves
- Laboratorio de Toxicología Ambiental, Facultad de Química, Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan s/n, Col. Residencial Colón, 50120, Toluca, Estado de México, Mexico
| | - Itzayana Pérez-Alvarez
- Facultad de Medicina, Universidad Autónoma del Estado de México. Paseo Tollocan /Jesús Carranza s/n. Toluca, 50120, Toluca, Estado de México, Mexico
| | - Diana Belén Onofre-Camarena
- Laboratorio de Toxicología Ambiental, Facultad de Química, Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan s/n, Col. Residencial Colón, 50120, Toluca, Estado de México, Mexico
| | - Verónica Margarita Gutiérrez-Noya
- Laboratorio de Toxicología Ambiental, Facultad de Química, Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan s/n, Col. Residencial Colón, 50120, Toluca, Estado de México, Mexico
| | - Karina Elisa Rosales-Pérez
- Laboratorio de Toxicología Ambiental, Facultad de Química, Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan s/n, Col. Residencial Colón, 50120, Toluca, Estado de México, Mexico
| | - José Manuel Orozco-Hernández
- Laboratorio de Toxicología Ambiental, Facultad de Química, Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan s/n, Col. Residencial Colón, 50120, Toluca, Estado de México, Mexico
| | - María Dolores Hernández-Navarro
- Laboratorio de Toxicología Ambiental, Facultad de Química, Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan s/n, Col. Residencial Colón, 50120, Toluca, Estado de México, Mexico
| | - Hariz Islas Flores
- Laboratorio de Toxicología Ambiental, Facultad de Química, Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan s/n, Col. Residencial Colón, 50120, Toluca, Estado de México, Mexico
| | - Leobardo Manuel Gómez-Olivan
- Laboratorio de Toxicología Ambiental, Facultad de Química, Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan s/n, Col. Residencial Colón, 50120, Toluca, Estado de México, Mexico.
| |
Collapse
|
4
|
Haskell-Mendoza AP, Srinivasan ES, Gonzalez AT, Reason EH, Jackson JD, Flusche AM, Wachsmuth LP, Lerner E, Underwood D, Buckley ED, Zaidi SE, Herndon JE, Fecci PE. Combination checkpoint blockade and laser interstitial thermal therapy in radiographically progressive non-small cell lung cancer brain metastases. Neurooncol Adv 2024; 6:vdae207. [PMID: 39717437 PMCID: PMC11664258 DOI: 10.1093/noajnl/vdae207] [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] [Indexed: 12/25/2024] Open
Abstract
Background Laser interstitial thermal therapy (LITT) is a minimally invasive surgical treatment being employed frequently for radiographically progressive brain metastases. Considerable interest exists in combining LITT-mediated in situ vaccination to license immune checkpoint blockade (ICB). No studies have examined the clinical feasibility of this combination in brain metastases. Methods All patients receiving LITT for radiographically progressive non-small cell lung carcinoma (NSCLC) brain metastases at a single center from 2015 to 2023 were retrospectively reviewed. Combination therapy was defined as ICB within 6 weeks of LITT. Clinical data, post-LITT freedom from local progression, and overall survival (OS) were collected. Adverse events (AEs) were evaluated according to Common Terminology Criteria. Results Eighteen patients received LITT + ICB for a total of 19 lesions. The median time between therapies was 2.29 weeks (range 0.85-5.98). In comparison to NSCLC patients receiving LITT alone or with targeted therapy (LITT only) (n = 25), there was no decrement in procedural outcomes. Patients receiving LITT + ICB discontinued steroids at a median of 11 (4-147) days post-LITT vs. 24 (3-242) days for patients receiving LITT only (P = .62). At study cutoff, the local control rate was 18/19 (94.7%) lesions in the LITT + ICB group and 22/25 (88.0%) in the LITT only group. There were 3 and 5 AEs ≥Grade 3 in the LITT + ICB and LITT-only groups, respectively. Conclusions Combination of LITT and ICB does not compromise procedural outcomes or time to steroid discontinuation in NSCLC. Prospective studies are needed to assess biomarkers of immune response.
Collapse
Affiliation(s)
| | - Ethan S Srinivasan
- Department of Neurosurgery, Johns Hopkins University Hospital, Baltimore, Maryland, USA
| | | | - Ellery H Reason
- Duke University School of Medicine, Durham, North Carolina, USA
| | - Joshua D Jackson
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USA
| | | | - Lucas P Wachsmuth
- Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Emily Lerner
- Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Delaney Underwood
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Evan D Buckley
- Biostatistics Shared Resource, Duke Cancer Institute, Durham, North Carolina, USA
| | - Saif E Zaidi
- Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USA
| | - James E Herndon
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Peter E Fecci
- Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
5
|
Mistry AM. Perioperative dexamethasone in high-grade gliomas: the short-term benefits and long-term harms. Front Oncol 2023; 13:1335730. [PMID: 38162484 PMCID: PMC10755919 DOI: 10.3389/fonc.2023.1335730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 12/04/2023] [Indexed: 01/03/2024] Open
Abstract
Dexamethasone has been commonly given to patients with a presumed new GBM in relatively large doses (6-16 mg daily for 1-2 weeks) since the 1960s without any rigorous evidence. This treatment with dexamethasone before the diagnosis and adjuvant therapy makes GBM patients unique compared to other newly diagnosed cancer patients. While dexamethasone may be beneficial, recent studies suggest that this potent immunosuppressant with pleiotropic effects is harmful in the long term. This perspective article summarizes the disadvantages of perioperative dexamethasone from multiple facets. It concludes that these growing data mandate rigorously testing the benefits of using perioperative dexamethasone.
Collapse
Affiliation(s)
- Akshitkumar M. Mistry
- Department of Neurological Surgery, University of Louisville, Louisville, KY, United States
| |
Collapse
|
6
|
Zhang I, Liu S, Zhang L, Liang R, Fang Q, Zhao J, Ren L, Medina E, Filippov A, Bonjoc KJ, Chaudhry A, Dayyani M, Bild A, Badie B. RAGE ablation attenuates glioma progression and enhances tumor immune responses by suppressing galectin-3 expression. Neuro Oncol 2023; 25:886-898. [PMID: 36394567 PMCID: PMC10158202 DOI: 10.1093/neuonc/noac250] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Malignant gliomas consist of heterogeneous cellular components that have adopted multiple overlapping escape mechanisms that overcome both targeted and immune-based therapies. The receptor for advanced glycation end products (RAGE) is a member of the immunoglobulin superfamily that is activated by diverse proinflammatory ligands present in the tumor microenvironment. Activation of RAGE by its ligands stimulates multiple signaling pathways that are important in tumor growth and invasion. However, treatment strategies that only target the interaction of RAGE with its ligands are ineffective as cancer therapies due to the abundance and diversity of exogenous RAGE ligands in gliomas. METHODS As an alternative approach to RAGE ligand inhibition, we evaluated the genetic ablation of RAGE on the tumorigenicity of 2 syngeneic murine glioma models. RAGE expression was inhibited in the GL261 and K-Luc gliomas by shRNA and CRSPR/Cas9 techniques prior to intracranial implantation. Tumor growth, invasion, and inflammatory responses were examined by histology, survival, Nanostring, and flow cytometry. RESULTS Intracellular RAGE ablation abrogated glioma growth and invasion by suppressing AKT and ERK1/2 activities and by downregulating MMP9 expression. Interestingly, RAGE inhibition in both glioma models enhanced tumor inflammatory responses by downregulating the expression of galectin-3 and potentiated immunotherapy responses to immune checkpoint blockade. CONCLUSIONS We demonstrated that intracellular RAGE ablation suppresses multiple cellular pathways that are important in glioma progression, invasion, and immune escape. These findings strongly support the development of RAGE ablation as a treatment strategy for malignant gliomas.
Collapse
Affiliation(s)
- Ian Y Zhang
- Division of Neurosurgery, City of Hope Beckman Research Institute, Duarte, California, USA
| | - Shunan Liu
- Department of Pharmacology, The Pharmacy School of Jilin University, Changchun, Jilin Province, P.R. China
| | - Leying Zhang
- Division of Neurosurgery, City of Hope Beckman Research Institute, Duarte, California, USA
| | - Rongrui Liang
- Department of Oncology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, Jiangsu, China
| | - Qingxiao Fang
- Colorectal Cancer Surgery Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jie Zhao
- Tianjin Union Medicine Center, Tianjin, 300123, P.R. China
| | - Lyuzhi Ren
- Division of Neurosurgery, City of Hope Beckman Research Institute, Duarte, California, USA
| | - Eric F Medina
- Graduate Education Progr-BRI, City of Hope Beckman Research Institute, Duarte, California, USA
| | - Aleksandr Filippov
- Graduate Education Progr-BRI, City of Hope Beckman Research Institute, Duarte, California, USA
| | - Kimberley-Jane Bonjoc
- Imaging Administration, City of Hope Beckman Research Institute, Duarte, California, USA
| | - Ammar Chaudhry
- Diagnostic Radiology, City of Hope Beckman Research Institute, Duarte, California, USA
| | - Mojtaba Dayyani
- Division of Neurosurgery, City of Hope Beckman Research Institute, Duarte, California, USA
| | - Andrea H Bild
- Medical Oncology, City of Hope Beckman Research Institute, Duarte, California, USA
| | - Behnam Badie
- Division of Neurosurgery, City of Hope Beckman Research Institute, Duarte, California, USA
| |
Collapse
|
7
|
Valiente M, Sepúlveda JM, Pérez A. Emerging targets for cancer treatment: S100A9/RAGE. ESMO Open 2023; 8:100751. [PMID: 36652782 PMCID: PMC9860424 DOI: 10.1016/j.esmoop.2022.100751] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/24/2022] [Accepted: 12/05/2022] [Indexed: 01/19/2023] Open
Abstract
Developing better treatments that work for the majority of patients with brain metastasis (BM) is highly necessary. Complementarily, avoiding those therapeutic procedures that will not benefit a specific patient is also very relevant. In general, existing therapies for patients with BM could be improved in terms of molecular stratification and therapeutic efficacy. By questioning the benefit of whole brain radiotherapy as provided nowadays and the lack of biomarkers detecting radioresistance, we identified S100A9 and receptor for advanced glycation end-products (RAGE) as a liquid biopsy biomarker and a potential target for a radiosensitizer, respectively. Both of them are being clinically tested as part of the first comprehensive molecular strategy to personalized radiotherapy in BM.
Collapse
Affiliation(s)
| | - J M Sepúlveda
- Neuro-Oncology Unit, Hospital Universitario 12 de Octubre, Madrid; Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid.
| | - A Pérez
- Neuro-Oncology Unit, Hospital Universitario 12 de Octubre, Madrid; Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid; Servicio de Neurocirugía, Hospital Universitario 12 de Octubre, Madrid; Departamento de Cirugía, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain.
| |
Collapse
|