1
|
Osa-Andrews B, van Wijk XMR, Herrera Rivera N, Seifert RP, Harris NS, Marin MJ. An Introduction to the Complete Blood Count for Clinical Chemists: White Blood Cells. J Appl Lab Med 2025; 10:459-475. [PMID: 39873240 DOI: 10.1093/jalm/jfaf004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 12/26/2024] [Indexed: 01/30/2025]
Abstract
BACKGROUND The most frequently ordered laboratory test worldwide is the complete blood count (CBC). As clinical chemists are increasingly assigned to assist or direct laboratories outside of the traditional clinical chemistry sections, such as the automated hematology section, expertise must be established. This review article is a dedication to that ongoing effort. CONTENT In this primer, the white blood cell (WBC) test components of the CBC are introduced, followed by a discussion of the laboratory evaluation of leukopenia and leukocytosis. SUMMARY The laboratorian's approach to consult cases should be guided by the patient's clinical history and presentation while being able to provide key laboratory-based insights to assist in resolving result discrepancies that may otherwise go unnoticed.
Collapse
Affiliation(s)
- Bremansu Osa-Andrews
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, United States
| | - Xander M R van Wijk
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, United States
| | | | - Robert P Seifert
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, United States
| | - Neil S Harris
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, United States
| | - Maximo J Marin
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
2
|
Dasanu CA, Plaxe SC. Temozolomide overcoming resistance to immune checkpoint inhibitors in relapsed/refractory metastatic melanoma? Insights from a single center series. J Oncol Pharm Pract 2025; 31:321-324. [PMID: 39584741 DOI: 10.1177/10781552241302421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
There is a need to develop more effective salvage therapies for patients with relapsed melanoma of the skin. Research has shown that chemotherapy-induced cancer cell death may increase immunogenic antigen exposure, or upregulation of co-inhibitory ligands such as PD-L1, thereby augmenting immune checkpoint inhibitor (ICI) efficacy. In addition, chemotherapy preconditioning may lead to depletion of Tregs, known to suppress immune anti-melanoma responses. As a result, regimens including both chemotherapy and ICI constructs are currently successfully employed in the 1st line therapy of many solid tumors. We report a series of three patients with metastatic melanoma, refractory to ICI treatment, who responded to salvage therapy with temozolomide (TMZ) in combination with PD-1 inhibitors, with or without CTLA-4 inhibitors. The responses were durable, each lasting more than 12 months. In two patients, complete responses are ongoing at 13 and 15 months, respectively. Randomized clinical trials with TMZ plus ICIs for patients with relapsed or refractory malignant melanoma seem warranted.
Collapse
Affiliation(s)
- Constantin A Dasanu
- Lucy Curci Cancer Center, Eisenhower Health, Rancho Mirage, CA, USA
- Department of Medical Oncology, UC San Diego Health System, San Diego, CA, USA
| | - Steven C Plaxe
- Lucy Curci Cancer Center, Eisenhower Health, Rancho Mirage, CA, USA
- Department of Medical Oncology, UC San Diego Health System, San Diego, CA, USA
| |
Collapse
|
3
|
Deng C, Yang S, Pu C, Bai X, Tian C, Feng M. Temozolomide Treatment in Refractory Pituitary Adenomas and Pituitary Carcinomas. Neuroendocrinology 2025; 115:335-350. [PMID: 39778549 PMCID: PMC11991747 DOI: 10.1159/000543427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 01/03/2025] [Indexed: 01/11/2025]
Abstract
BACKGROUND Temozolomide (TMZ), a nonclassical alkylating agent, possesses lipophilic properties that allow it to cross the blood-brain barrier, making it active within the central nervous system. Furthermore, the adverse reactions of the TMZ are relatively mild, which is why it is currently recommended as a first-line chemotherapy drug for refractory pituitary adenomas (RPAs) and pituitary carcinomas (PCs). SUMMARY Systematic evaluations indicate a radiological response rate of 41% and a hormonal response rate of 53%, underscoring TMZ clinical efficacy, particularly when combined with radiotherapy. Functional tumors demonstrate a higher response rate compared to nonfunctional tumors. While the optimal duration of TMZ treatment remains undetermined, studies suggest that longer therapy durations may lead to better prognoses. Additionally, prior to TMZ administration, it is advisable to conduct immunohistochemical analysis of O6-methylguanine-DNA methyltransferase, MSH2, MSH6, MLH1, PMS2, and N-methylpurine DNA glycosylase to assess the potential impact of repair mechanisms such as direct repair, mismatch repair pathway, and base excision repair on TMZ treatment. The efficacy of TMZ analogs, combined TMZ therapies, and TMZ with nanomaterials following TMZ treatment failure remains uncertain. KEY MESSAGES The involvement of experienced multidisciplinary pituitary teams in all management decisions for RPAs/PCs patients is essential.
Collapse
Affiliation(s)
- Congcong Deng
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Shuangjian Yang
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Changqin Pu
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xuexue Bai
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Chenxin Tian
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Ming Feng
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
4
|
Fasano M, Pirozzi M, De Falco V, Miceli CC, Farese S, Zotta A, Famiglietti V, Vitale P, Di Giovanni I, Brancati C, Carfora V, Solari D, Somma T, Cavallo LM, Cappabianca P, Conson M, Pacelli R, Ciardiello F, Addeo R. Temozolomide based treatment in glioblastoma: 6 vs. 12 months. Oncol Lett 2024; 28:418. [PMID: 39006948 PMCID: PMC11240269 DOI: 10.3892/ol.2024.14551] [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: 09/12/2023] [Accepted: 11/28/2023] [Indexed: 07/16/2024] Open
Abstract
The Stupp regimen remains the standard treatment for newly diagnosed glioblastomas, although the prognosis remains poor. Several temozolomide alternative schedules have been studied, with extended adjuvant treatment (>6 cycles of temozolomide) frequently used, although different trials have indicated contrasting results. Survival data of 87 patients who received 6 ('6C' group) or 12 ('12C' group) cycles of temozolomide were collected between 2012 and 2022. A total of 45 patients were included in the 6C group and 42 patients were included in the 12C group. Data on isocitrate dehydrogenase mutation and methylguanine-DNA-methyltransferase (MGMT) promoter methylation status were also collected. The 12C group exhibited statistically significantly improved overall survival [OS; 22.8 vs. 17.5 months; hazard ratio (HR), 0.47; 95% CI, 0.30-0.73; P=0.001] and progression-free survival (15.3 vs. 9 months; HR, 0.39; 95% CI, 0.25-0.62; P=0.001). However, in the subgroup analysis according to MGMT status, OS in the 12C group was significantly superior to OS in the 6C group only in the MGMT unmethylated tumors. The present data suggested that extended adjuvant temozolomide appeared to be more effective than the conventional six cycles.
Collapse
Affiliation(s)
- Morena Fasano
- Medical Oncology Unit, Department of Precision Medicine, University of Campania Luigi Vanvitelli, I-80131 Naples, Italy
| | - Mario Pirozzi
- Medical Oncology Unit, Department of Precision Medicine, University of Campania Luigi Vanvitelli, I-80131 Naples, Italy
| | - Vincenzo De Falco
- Oncology Unit, 'San Giovanni di Dio' Hospital, ASL Napoli 2 Nord, I-80020 Frattamaggiore, Italy
| | - Chiara Carmen Miceli
- Medical Oncology Unit, Department of Precision Medicine, University of Campania Luigi Vanvitelli, I-80131 Naples, Italy
| | - Stefano Farese
- Medical Oncology Unit, Department of Precision Medicine, University of Campania Luigi Vanvitelli, I-80131 Naples, Italy
| | - Alessia Zotta
- Medical Oncology Unit, Department of Precision Medicine, University of Campania Luigi Vanvitelli, I-80131 Naples, Italy
| | - Vincenzo Famiglietti
- Medical Oncology Unit, Department of Precision Medicine, University of Campania Luigi Vanvitelli, I-80131 Naples, Italy
| | - Pasquale Vitale
- Oncology Unit, 'San Giovanni di Dio' Hospital, ASL Napoli 2 Nord, I-80020 Frattamaggiore, Italy
| | - Ilaria Di Giovanni
- Oncology Unit, 'San Giovanni di Dio' Hospital, ASL Napoli 2 Nord, I-80020 Frattamaggiore, Italy
| | - Christian Brancati
- Oncology Unit, 'San Giovanni di Dio' Hospital, ASL Napoli 2 Nord, I-80020 Frattamaggiore, Italy
| | - Vincenzo Carfora
- Radiation Oncology Unit, Department of Radiation Oncology, 'San Pio' Hospital, I-82100 Benevento, Italy
| | - Domenico Solari
- Division of Neurosurgery, Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, I-80131 Naples, Italy
| | - Teresa Somma
- Division of Neurosurgery, Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, I-80131 Naples, Italy
| | - Luigi Maria Cavallo
- Division of Neurosurgery, Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, I-80131 Naples, Italy
| | - Paolo Cappabianca
- Division of Neurosurgery, Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, I-80131 Naples, Italy
| | - Manuel Conson
- Department of Advanced Biomedical Sciences, University of Naples Federico II, I-80131 Naples, Italy
| | - Roberto Pacelli
- Department of Advanced Biomedical Sciences, University of Naples Federico II, I-80131 Naples, Italy
| | - Fortunato Ciardiello
- Medical Oncology Unit, Department of Precision Medicine, University of Campania Luigi Vanvitelli, I-80131 Naples, Italy
| | - Raffaele Addeo
- Oncology Unit, 'San Giovanni di Dio' Hospital, ASL Napoli 2 Nord, I-80020 Frattamaggiore, Italy
| |
Collapse
|
5
|
Stergiopoulos GM, Concilio SC, Galanis E. An Update on the Clinical Status, Challenges, and Future Directions of Oncolytic Virotherapy for Malignant Gliomas. Curr Treat Options Oncol 2024; 25:952-991. [PMID: 38896326 PMCID: PMC11878440 DOI: 10.1007/s11864-024-01211-6] [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] [Accepted: 04/24/2024] [Indexed: 06/21/2024]
Abstract
OPINION STATEMENT Malignant gliomas are common central nervous system tumors that pose a significant clinical challenge due to the lack of effective treatments. Glioblastoma (GBM), a grade 4 malignant glioma, is the most prevalent primary malignant brain tumor and is associated with poor prognosis. Current clinical trials are exploring various strategies to combat GBM, with oncolytic viruses (OVs) appearing particularly promising. In addition to ongoing and recently completed clinical trials, one OV (Teserpaturev, Delytact®) received provisional approval for GBM treatment in Japan. OVs are designed to selectively target and eliminate cancer cells while promoting changes in the tumor microenvironment that can trigger and support long-lasting anti-tumor immunity. OVs offer the potential to remodel the tumor microenvironment and reverse systemic immune exhaustion. Additionally, an increasing number of OVs are armed with immunomodulatory payloads or combined with immunotherapy approaches in an effort to promote anti-tumor responses in a tumor-targeted manner. Recently completed oncolytic virotherapy trials can guide the way for future treatment individualization through patient preselection, enhancing the likelihood of achieving the highest possible clinical success. These trials also offer valuable insight into the numerous challenges inherent in malignant glioma treatment, some of which OVs can help overcome.
Collapse
Affiliation(s)
| | | | - Evanthia Galanis
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA.
- Department of Oncology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
6
|
Zhou J, Zheng H, Zhang H, Yu W, Li B, Ye L, Wang L. MCM5 is a Novel Therapeutic Target for Glioblastoma. Onco Targets Ther 2024; 17:371-381. [PMID: 38765057 PMCID: PMC11100520 DOI: 10.2147/ott.s457600] [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: 03/18/2024] [Accepted: 05/08/2024] [Indexed: 05/21/2024] Open
Abstract
Objective MCM5 is a DNA licensing factor involved in cell proliferation and has been previously established as an excellent biomarker in a number of malignancies. Nevertheless, the role of MCM5 in GBM has not been fully clarified. The present study aimed to investigate the potential roles of MCM5 in the treatment of GBM and to elucidate its underlying mechanism, which is beneficial for developing new therapeutic strategies and predicting prognosis. Methods Firstly, we obtained transcriptomic and proteomic data from the TCGA and CPTAC databases on glioma patients. Employing the DeSeq2 R package, we then identified genes with joint differential expression in GBM tissues subjected to chemotherapy. To develop a prognostic risk score model, we performed univariate and multivariate Cox regression analyses. In vitro knockdown and overexpression of MCM5 were used to further investigate the biological functions of GBM cells. Additionally, we also delved into the upstream regulation of MCM5, revealing associations with several transcription factors. Finally, we investigated differences in immune cell infiltration and drug sensitivity across diverse risk groups identified in the prognostic risk model. Results In this study, the chemotherapy-treated GBM samples exhibited consistent alterations in 46 upregulated and 94 downregulated genes at both the mRNA and protein levels. Notably, MCM5 emerged as a gene with prognostic significance as well as potential therapeutic relevance. In vitro experiments subsequently validated the role of increased MCM5 expression in promoting GBM cell proliferation and resistance to TMZ. Correlations with transcription factors such as CREB1, CTCF, NFYB, NRF1, PBX1, TEAD1, and USF1 were discovered during upstream regulatory analysis, enriching our understanding of MCM5 regulatory mechanisms. The study additionally delves into immune cell infiltration and drug sensitivity, providing valuable insights for personalized treatment approaches. Conclusion This study identifies MCM5 as a key player in GBM, demonstrating its prognostic significance and potential therapeutic relevance by elucidating its role in promoting cell proliferation and resistance to chemotherapy.
Collapse
Affiliation(s)
- Jian Zhou
- Hyperbaric Oxygen Department, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Medical School, Shenzhen, 518055, People’s Republic of China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, People’s Republic of China
| | - Housheng Zheng
- Hyperbaric Oxygen Department, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Medical School, Shenzhen, 518055, People’s Republic of China
| | - Huiru Zhang
- Hyperbaric Oxygen Department, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Medical School, Shenzhen, 518055, People’s Republic of China
| | - Wenqiang Yu
- Hyperbaric Oxygen Department, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Medical School, Shenzhen, 518055, People’s Republic of China
| | - Baoer Li
- Hyperbaric Oxygen Department, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Medical School, Shenzhen, 518055, People’s Republic of China
| | - Liang Ye
- Hyperbaric Oxygen Department, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Medical School, Shenzhen, 518055, People’s Republic of China
| | - Lu Wang
- Hyperbaric Oxygen Department, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Medical School, Shenzhen, 518055, People’s Republic of China
| |
Collapse
|
7
|
Bhati V, Kumar A, Lather V, Sharma R, Pandita D. Association of temozolomide with progressive multifocal leukoencephalopathy: a disproportionality analysis integrated with network pharmacology. Expert Opin Drug Saf 2024; 23:649-658. [PMID: 37915230 DOI: 10.1080/14740338.2023.2278682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 09/15/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND Temozolomide (TMZ) is an alkylating agent approved for the management of glioblastoma. The TMZ is not known for progressive multifocal leukoencephalopathy (PML). The main objective of the current study is to find out the association of TMZ with PML using disproportionality analysis of FDA Adverse Event Reporting System (FAERS) data integrated with network pharmacological approaches. RESEARCH DESIGN AND METHODS OpenVigil tool was used to query the FAERS database. The disproportionality measures were calculated. The network has been constructed using Cytoscape. Finally, the possible binding interactions were studied using Glide, Schrödinger Inc. RESULTS A total number of 3502 cases of PML were reported in the FAERS database. Out of these, 10 cases were found with TMZ. The subgroup analysis results have shown a greater number of cases in females. The network has indicated the involvement of human mitogen-activated protein kinase, 3-phosphoinositide-dependent protein kinase 1 protein, human mTOR complex protein, phosphatidylinositol 4,5-bisphosphate 3-kinase protein, and glycogen synthase kinase-3 beta protein. The docking results have indicated good interactions of TMZ with active site of glycogen synthase kinase-3 beta and mitogen-activated protein kinase 1 as compared to other identified targets. CONCLUSION The PML is identified as novel signal with temozolomide.
Collapse
Affiliation(s)
- Vipin Bhati
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| | - Anoop Kumar
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| | - Viney Lather
- Centre for Pharmaceutical Chemistry and Analysis, Amity Institute of Pharmacy, Amity University Uttar Pradesh, Noida, India
| | - Ruchika Sharma
- Centre for Precision Medicine and Pharmacy, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, India
| | - Deepti Pandita
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences and Research, New Delhi, India
| |
Collapse
|
8
|
de Kermenguy F, Benzazon N, Maury P, Vauclin R, M'hamdi M, Cifliku V, Limkin E, Diallo I, Morel D, Milewski C, Clémenson C, Mondini M, Deutsch E, Robert C. LymphoDose: a lymphocyte dose estimation framework-application to brain radiotherapy. Phys Med Biol 2024; 69:105009. [PMID: 38593817 DOI: 10.1088/1361-6560/ad3c8d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 04/09/2024] [Indexed: 04/11/2024]
Abstract
Objective. Severe radiation-induced lymphopenia occurs in 40% of patients treated for primary brain tumors and is an independent risk factor of poor survival outcomes. We developed anin-silicoframework that estimates the radiation doses received by lymphocytes during volumetric modulated arc therapy brain irradiation.Approach. We implemented a simulation consisting of two interconnected compartmental models describing the slow recirculation of lymphocytes between lymphoid organs (M1) and the bloodstream (M2). We used dosimetry data from 33 patients treated with chemo-radiation for glioblastoma to compare three cases of the model, corresponding to different physical and biological scenarios: (H1) lymphocytes circulation only in the bloodstream i.e. circulation inM2only; (H2) lymphocytes recirculation between lymphoid organs i.e. circulation inM1andM2interconnected; (H3) lymphocytes recirculation between lymphoid organs and deep-learning computed out-of-field (OOF) dose to head and neck (H&N) lymphoid structures. A sensitivity analysis of the model's parameters was also performed.Main results. For H1, H2 and H3 cases respectively, the irradiated fraction of lymphocytes was 99.8 ± 0.7%, 40.4 ± 10.2% et 97.6 ± 2.5%, and the average dose to irradiated pool was 309.9 ± 74.7 mGy, 52.6 ± 21.1 mGy and 265.6 ± 48.5 mGy. The recirculation process considered in the H2 case implied that irradiated lymphocytes were irradiated in the field only 1.58 ± 0.91 times on average after treatment. The OOF irradiation of H&N lymphoid structures considered in H3 was an important contribution to lymphocytes dose. In all cases, the estimated doses are low compared with lymphocytes radiosensitivity, and other mechanisms could explain high prevalence of RIL in patients with brain tumors.Significance. Our framework is the first to take into account OOF doses and recirculation in lymphocyte dose assessment during brain irradiation. Our results demonstrate the need to clarify the indirect effects of irradiation on lymphopenia, in order to potentiate the combination of radio-immunotherapy or the abscopal effect.
Collapse
Affiliation(s)
- François de Kermenguy
- Université Paris-Saclay, Gustave Roussy, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, F-94800, Villejuif, France
| | - Nathan Benzazon
- Université Paris-Saclay, Gustave Roussy, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, F-94800, Villejuif, France
| | - Pauline Maury
- Université Paris-Saclay, Gustave Roussy, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, F-94800, Villejuif, France
- Gustave Roussy, Département de radiothérapie, F-94800, Villejuif, France
| | | | - Meissane M'hamdi
- Université Paris-Saclay, Gustave Roussy, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, F-94800, Villejuif, France
| | - Vjona Cifliku
- Université Paris-Saclay, Gustave Roussy, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, F-94800, Villejuif, France
| | - Elaine Limkin
- Université Paris-Saclay, Gustave Roussy, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, F-94800, Villejuif, France
- Gustave Roussy, Département de radiothérapie, F-94800, Villejuif, France
| | - Ibrahima Diallo
- Université Paris-Saclay, Gustave Roussy, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, F-94800, Villejuif, France
| | - Daphné Morel
- Université Paris-Saclay, Gustave Roussy, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, F-94800, Villejuif, France
| | - Candice Milewski
- Gustave Roussy, Département de radiothérapie, F-94800, Villejuif, France
| | - Céline Clémenson
- Université Paris-Saclay, Gustave Roussy, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, F-94800, Villejuif, France
| | - Michele Mondini
- Université Paris-Saclay, Gustave Roussy, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, F-94800, Villejuif, France
| | - Eric Deutsch
- Université Paris-Saclay, Gustave Roussy, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, F-94800, Villejuif, France
- Gustave Roussy, Département de radiothérapie, F-94800, Villejuif, France
| | - Charlotte Robert
- Université Paris-Saclay, Gustave Roussy, Inserm U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, F-94800, Villejuif, France
- Gustave Roussy, Département de radiothérapie, F-94800, Villejuif, France
| |
Collapse
|
9
|
Kim J, Choi H, Jeun SS, Ahn S. From lymphopenia to restoration: IL-7 immunotherapy for lymphocyte recovery in glioblastoma. Cancer Lett 2024; 588:216714. [PMID: 38369003 DOI: 10.1016/j.canlet.2024.216714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/30/2024] [Accepted: 02/03/2024] [Indexed: 02/20/2024]
Abstract
Glioblastoma, the most prevalent malignant primary brain tumor, presents substantial treatment challenges because of its inherent aggressiveness and limited therapeutic options. Lymphopenia, defined as reduced peripheral blood lymphocyte count, commonly occurs as a consequence of the disease and its treatment. Recent studies have associated lymphopenia with a poor prognosis. Factors that contribute to lymphopenia include radiotherapy, chemotherapy, and the tumor itself. Patients who are female, older, using dexamethasone, or receiving higher doses of radiation therapy are particularly vulnerable to this condition. Several preclinical studies have explored the use of interleukin-7, a crucial cytokine for lymphocyte homeostasis, to restore lymphocyte counts and potentially rebuild the immune system to combat glioblastoma cells. With the development of recombinant interleukin-7 for prolonged activity in the body, various clinical trials are underway to explore this treatment in patients with glioblastoma. Our study provides a comprehensive summary of the incidence of lymphopenia, its potential biological background, and the associated clinical risk factors. Furthermore, we reviewed several clinical trials using IL-7 cytokine therapy in glioblastoma patients. We propose IL-7 as a promising immunotherapeutic strategy for glioblastoma treatment. We are optimistic that our study will enhance understanding of the complex interplay between lymphopenia and glioblastoma and will pave the way for the development of more effective treatment modalities.
Collapse
Affiliation(s)
- Joonseok Kim
- College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Haeyoun Choi
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sin-Soo Jeun
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Stephen Ahn
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
10
|
Lee HN, Do KH, Kim EY, Choe J, Sung H, Choi SH, Kim HJ. Comparative Analysis of CT Findings and Clinical Outcomes in Adult Patients With Disseminated and Localized Pulmonary Nocardiosis. J Korean Med Sci 2024; 39:e107. [PMID: 38529577 DOI: 10.3346/jkms.2024.39.e107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/01/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Pulmonary nocardiosis is a rare opportunistic infection with occasional systemic dissemination. This study aimed to investigate the computed tomography (CT) findings and prognosis of pulmonary nocardiosis associated with dissemination. METHODS We conducted a retrospective analysis of patients diagnosed with pulmonary nocardiosis between March 2001 and September 2023. We reviewed the chest CT findings and categorized them based on the dominant CT findings as consolidation, nodules and/or masses, consolidation with multiple nodules, and nodular bronchiectasis. We compared chest CT findings between localized and disseminated pulmonary nocardiosis and identified significant prognostic factors associated with 12-month mortality using multivariate Cox regression analysis. RESULTS Pulmonary nocardiosis was diagnosed in 75 patients, of whom 14 (18.7%) had dissemination, including involvement of the brain in 9 (64.3%) cases, soft tissue in 3 (21.4%) cases and positive blood cultures in 3 (21.4%) cases. Disseminated pulmonary nocardiosis showed a higher frequency of cavitation (64.3% vs. 32.8%, P = 0.029) and pleural effusion (64.3% vs. 29.5%, P = 0.014) compared to localized infection. The 12-month mortality rate was 25.3%. The presence of dissemination was not a significant prognostic factor (hazard ratio [HR], 0.80; confidence interval [CI], 0.23-2.75; P = 0.724). Malignancy (HR, 9.73; CI, 2.32-40.72; P = 0.002), use of steroid medication (HR, 3.72; CI, 1.33-10.38; P = 0.012), and a CT pattern of consolidation with multiple nodules (HR, 4.99; CI, 1.41-17.70; P = 0.013) were associated with higher mortality rates. CONCLUSION Pulmonary nocardiosis with dissemination showed more frequent cavitation and pleural effusion compared to cases without dissemination, but dissemination alone did not affect the mortality rate of pulmonary nocardiosis.
Collapse
Affiliation(s)
- Han Na Lee
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyung-Hyun Do
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| | - Eun Young Kim
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jooae Choe
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Heungsup Sung
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sang-Ho Choi
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hwa Jung Kim
- Department of Clinical Epidemiology and Biostatistics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
11
|
Stepanenko AA, Sosnovtseva AO, Valikhov MP, Chernysheva AA, Abramova OV, Naumenko VA, Chekhonin VP. The need for paradigm shift: prognostic significance and implications of standard therapy-related systemic immunosuppression in glioblastoma for immunotherapy and oncolytic virotherapy. Front Immunol 2024; 15:1326757. [PMID: 38390330 PMCID: PMC10881776 DOI: 10.3389/fimmu.2024.1326757] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/23/2024] [Indexed: 02/24/2024] Open
Abstract
Despite significant advances in our knowledge regarding the genetics and molecular biology of gliomas over the past two decades and hundreds of clinical trials, no effective therapeutic approach has been identified for adult patients with newly diagnosed glioblastoma, and overall survival remains dismal. Great hopes are now placed on combination immunotherapy. In clinical trials, immunotherapeutics are generally tested after standard therapy (radiation, temozolomide, and steroid dexamethasone) or concurrently with temozolomide and/or steroids. Only a minor subset of patients with progressive/recurrent glioblastoma have benefited from immunotherapies. In this review, we comprehensively discuss standard therapy-related systemic immunosuppression and lymphopenia, their prognostic significance, and the implications for immunotherapy/oncolytic virotherapy. The effectiveness of immunotherapy and oncolytic virotherapy (viro-immunotherapy) critically depends on the activity of the host immune cells. The absolute counts, ratios, and functional states of different circulating and tumor-infiltrating immune cell subsets determine the net immune fitness of patients with cancer and may have various effects on tumor progression, therapeutic response, and survival outcomes. Although different immunosuppressive mechanisms operate in patients with glioblastoma/gliomas at presentation, the immunological competence of patients may be significantly compromised by standard therapy, exacerbating tumor-related systemic immunosuppression. Standard therapy affects diverse immune cell subsets, including dendritic, CD4+, CD8+, natural killer (NK), NKT, macrophage, neutrophil, and myeloid-derived suppressor cell (MDSC). Systemic immunosuppression and lymphopenia limit the immune system's ability to target glioblastoma. Changes in the standard therapy are required to increase the success of immunotherapies. Steroid use, high neutrophil-to-lymphocyte ratio (NLR), and low post-treatment total lymphocyte count (TLC) are significant prognostic factors for shorter survival in patients with glioblastoma in retrospective studies; however, these clinically relevant variables are rarely reported and correlated with response and survival in immunotherapy studies (e.g., immune checkpoint inhibitors, vaccines, and oncolytic viruses). Our analysis should help in the development of a more rational clinical trial design and decision-making regarding the treatment to potentially improve the efficacy of immunotherapy or oncolytic virotherapy.
Collapse
Affiliation(s)
- Aleksei A. Stepanenko
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N.I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Anastasiia O. Sosnovtseva
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Marat P. Valikhov
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N.I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Anastasia A. Chernysheva
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Olga V. Abramova
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Victor A. Naumenko
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Vladimir P. Chekhonin
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N.I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
12
|
Qiu Q, Chen S, He H, Chen J, Ding X, Wang D, Yang J, Guo P, Li Y, Kim J, Sheng J, Gao C, Yin B, Zheng S, Wang J. An injectable signal-amplifying device elicits a specific immune response against malignant glioblastoma. Acta Pharm Sin B 2023; 13:5091-5106. [PMID: 38045037 PMCID: PMC10692361 DOI: 10.1016/j.apsb.2023.06.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/04/2023] [Accepted: 06/05/2023] [Indexed: 12/05/2023] Open
Abstract
Despite exciting achievements with some malignancies, immunotherapy for hypoimmunogenic cancers, especially glioblastoma (GBM), remains a formidable clinical challenge. Poor immunogenicity and deficient immune infiltrates are two major limitations to an effective cancer-specific immune response. Herein, we propose that an injectable signal-amplifying nanocomposite/hydrogel system consisting of granulocyte-macrophage colony-stimulating factor and imiquimod-loaded antigen-capturing nanoparticles can simultaneously amplify the chemotactic signal of antigen-presenting cells and the "danger" signal of GBM. We demonstrated the feasibility of this strategy in two scenarios of GBM. In the first scenario, we showed that this simultaneous amplification system, in conjunction with local chemotherapy, enhanced both the immunogenicity and immune infiltrates in a recurrent GBM model; thus, ultimately making a cold GBM hot and suppressing postoperative relapse. Encouraged by excellent efficacy, we further exploited this signal-amplifying system to improve the efficiency of vaccine lysate in the treatment of refractory multiple GBM, a disease with limited clinical treatment options. In general, this biomaterial-based immune signal amplification system represents a unique approach to restore GBM-specific immunity and may provide a beneficial preliminary treatment for other clinically refractory malignancies.
Collapse
Affiliation(s)
- Qiujun Qiu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Sunhui Chen
- Department of Pharmacy, Fujian Provincial Hospital & Provincial Clinical Medical College of Fujian Medical University, Fuzhou 350001, China
| | - Huining He
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Jixiang Chen
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Xinyi Ding
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Dongdong Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Jiangang Yang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Pengcheng Guo
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Yang Li
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Jisu Kim
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Jianyong Sheng
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Chao Gao
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Bo Yin
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Shihao Zheng
- Department of Neurosurgery, Fujian Provincial Hospital & Provincial Clinical Medical College of Fujian Medical University, Fuzhou 350001, China
| | - Jianxin Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
- Institute of Materia Medica, Academy of Chinese and Western Integrative Medicine, Fudan University, Shanghai 201203, China
| |
Collapse
|
13
|
Lee DH, Kim HR, Keam B, Kato K, Kuboki Y, Gao H, Yovine A, Robbins SH, Ahn M. Safety and tolerability of first-line durvalumab with tremelimumab and chemotherapy in esophageal squamous cell carcinoma. Cancer Med 2023; 12:16066-16075. [PMID: 37489066 PMCID: PMC10469840 DOI: 10.1002/cam4.6260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/05/2023] [Accepted: 06/05/2023] [Indexed: 07/26/2023] Open
Abstract
BACKGROUND Advanced or metastatic esophageal squamous cell carcinoma (ESCC) is associated with poor prognosis; new first-line systemic treatment options are needed. Combining immuno-oncology therapies with standard chemotherapy may represent a promising approach for the treatment of solid tumors. Results from a Phase Ib study evaluating durvalumab with tremelimumab and chemotherapy in patients with advanced or metastatic ESCC are reported. METHODS Adults with advanced or metastatic ESCC who were candidates for first-line platinum-based chemotherapy received durvalumab 1500 mg (Day 1), tremelimumab 75 mg (Day 1), cisplatin 80 mg/m2 (Day 1) and 5-fluorouracil (5-FU) 800 mg/m2 (Days 1-5) in 28-day cycles until disease progression or discontinuation due to toxicity. The study consisted of safety run-in (Part A) and expansion (Part B) periods. The primary endpoint was safety. Antitumor activity was an exploratory endpoint. RESULTS Sixteen patients were enrolled, 6 in Part A and 10 in Part B, and received a median of 4.0 treatment cycles. All patients were Asian; median age was 65.0 years. All patients experienced adverse events (AEs) related to cisplatin and 5-FU, and 8 (50.0%) patients experienced AEs related to durvalumab and tremelimumab. Grade ≥3 treatment-related AEs occurred in 7 (43.8%) patients. There were no deaths associated with AEs. Six (37.5%) patients achieved an objective response. Median progression-free survival was 3.75 months, and median overall survival was 9.69 months. CONCLUSIONS Durvalumab with tremelimumab and chemotherapy demonstrated manageable safety and antitumor activity in patients with advanced or metastatic ESCC, warranting further investigation in randomized trials. Registered with ClinicalTrials.gov: NCT02658214.
Collapse
Affiliation(s)
- Dae Ho Lee
- Department of OncologyUniversity of Ulsan College of Medicine, Asan Medical CenterSeoulSouth Korea
| | - Hye Ryun Kim
- Division of Medical Oncology, Department of Internal MedicineYonsei Cancer Center, Yonsei University College of MedicineSeoulSouth Korea
| | - Bhumsuk Keam
- Department of Internal MedicineSeoul National University HospitalSeoulSouth Korea
| | - Ken Kato
- Department of Head and Neck, Esophageal OncologyNational Cancer Center HospitalTokyoJapan
| | - Yasutoshi Kuboki
- Gastrointestinal Oncology DivisionNational Cancer Center HospitalEast KashiwaJapan
| | | | | | | | - Myung‐Ju Ahn
- Division of Hematology‐Oncology, Department of MedicineSungkyunkwan University School of Medicine, Samsung Medical CenterSeoulSouth Korea
| |
Collapse
|
14
|
Dela Vega MP, Opinaldo PV, Batara JM. Differentiating Cerebral Toxoplasmosis and Tumor Recurrence by Thallium-201 Single-Photon Emission Computerized Tomography in a 28-Year-Old Female with Astrocytoma. Case Rep Oncol 2023; 16:357-362. [PMID: 37384212 PMCID: PMC10293939 DOI: 10.1159/000529830] [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: 12/17/2022] [Accepted: 02/09/2023] [Indexed: 06/30/2023] Open
Abstract
Cerebral toxoplasmosis is an opportunistic infection that, by itself, is difficult to differentiate from cerebral neoplasms by conventional neuroimaging. It rarely occurs concurrently in patients with a primary brain tumor but when it does, it makes diagnosis and management more difficult. This is a case of a 28-year-old female, diagnosed with a right frontal pleomorphic xanthoastrocytoma with several recurrences, treated with surgery, radiation, and chemotherapy. Three years from diagnosis, the patient was readmitted for generalized body weakness, fever, and a decrease in sensorium. A repeat cranial magnetic resonance imaging showed multiple enhancing lesions in both cerebral hemispheres and in the posterior fossa. Serum toxoplasma IgM and IgG antibody titers were elevated. Single-photon emission computerized tomography (SPECT) with thallium-201 did not show increased tracer uptake in these lesions, favoring toxoplasmosis over tumor recurrence. The patient was treated with trimethoprim-sulfamethoxazole with significant improvement. This is a rare account of cerebral toxoplasmosis arising in the setting of astrocytoma. This is also the first case report to demonstrate the value of thallium-201 SPECT in differentiating central nervous system infection from tumor recurrence, which is pivotal in management. More studies exploring the use of thallium-201 SPECT in distinguishing central nervous system infections from glioma and other malignant tumors should be undertaken to maximize this imaging modality in neuro-oncology practice.
Collapse
Affiliation(s)
- Mia Patrice Dela Vega
- Section of Adult Neurology, St. Luke's Medical Center, Institute for Neurosciences, Quezon City, Philippines
| | - Paul Vincent Opinaldo
- Section of Adult Neurology, St. Luke's Medical Center, Institute for Neurosciences, Quezon City, Philippines
| | - Julette Marie Batara
- Section of Adult Neurology, St. Luke's Medical Center, Institute for Neurosciences, Quezon City, Philippines
| |
Collapse
|
15
|
Abstract
Glioblastoma is the most aggressive primary brain tumor with a poor prognosis. The 2021 WHO CNS5 classification has further stressed the importance of molecular signatures in diagnosis although therapeutic breakthroughs are still lacking. In this review article, updates on the current and novel therapies in IDH-wildtype GBM will be discussed.
Collapse
Affiliation(s)
- Jawad M Melhem
- Division of Neurology, Department of Medicine, Faculty of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Jay Detsky
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Mary Jane Lim-Fat
- Division of Neurology, Department of Medicine, Faculty of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - James R Perry
- Division of Neurology, Department of Medicine, Faculty of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada.
| |
Collapse
|
16
|
Le Rhun E, Oppong FB, Vanlancker M, Stupp R, Nabors B, Chinot O, Wick W, Preusser M, Gorlia T, Weller M. Prognostic significance of therapy-induced myelosuppression in newly diagnosed glioblastoma. Neuro Oncol 2022; 24:1533-1545. [PMID: 35312789 PMCID: PMC9435483 DOI: 10.1093/neuonc/noac070] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Myelosuppression is the major toxicity encountered during temozolomide chemoradiotherapy for newly diagnosed glioblastoma. METHODS We assessed the association of myelosuppression (neutropenia, thrombocytopenia, anemia, and lymphopenia) during temozolomide chemoradiotherapy alone or in combination with experimental agents with progression-free survival (PFS) or overall survival (OS) in 2073 patients with newly diagnosed glioblastoma enrolled into five clinical trials: CENTRIC, CORE, EORTC 26082, AVAglio, and EORTC 26981. A landmark Cox model was used. For each primary association analysis, a significance level of 1.7% was used. RESULTS Lower neutrophil counts at baseline were associated with better PFS (P = .011) and OS (P < .001), independently of steroid intake. Females experienced uniformly more myelotoxicity than males. Lymphopenia during concomitant chemoradiotherapy was associated with OS (P = .009): low-grade (1-2) lymphopenia might be associated with superior OS (HR 0.78, 98.3% CI 0.58-1.06), whereas high-grade (3-4) lymphopenia might be associated with inferior OS (HR 1.08, 98.3% CI 0.75-1.54). There were no associations of altered hematological parameters during concomitant chemoradiotherapy with PFS. During maintenance chemoradiotherapy, no significant association was found between any parameter of myelosuppression and PFS or OS, although exploratory analysis at 5% significance level indicated that either mild-to-moderate (HR 0.76, 95% CI 0.62-0.93) or high-grade lymphopenia (HR 0.65, 95% CI 0.46-0.92) was associated with superior OS (P = .013), but not PFS. CONCLUSIONS The association of higher neutrophil counts at baseline with inferior PFS and OS requires further prospective evaluation. The link of therapy-induced lymphopenia to better outcome may guide the design for immunotherapy trials in newly diagnosed glioblastoma.
Collapse
Affiliation(s)
- Emilie Le Rhun
- Corresponding Author: Emilie Le Rhun, MD, PhD, Department of Neurosurgery, University Hospital Zurich, Frauenklinikstrasse 10, 8091 Zurich, Switzerland ()
| | | | | | - Roger Stupp
- Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
- Malnati Brain Tumor Center of the Lurie Comprehensive Cancer Center and Departments of Neursurgery and Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Burt Nabors
- Department of Neurology, Division of Neuro-Oncology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Olivier Chinot
- Aix-Marseille University, AP-HM, Service de Neuro-Oncologie, CHU Timone, Marseille, France
| | - Wolfgang Wick
- Department of Neurology and Neuro-oncology Program at the National Center for Tumor Diseases, University Hospital Heidelberg and German Cancer Research Center, Heidelberg, Germany
| | - Matthias Preusser
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | | | - Michael Weller
- Department of Neurology and Brain Tumor Center, University Hospital and University of Zurich, Zurich, Switzerland
| |
Collapse
|
17
|
Climans SA, Mason WP, Grunfeld E, Chan K. Clinical features of glioma patients who develop pneumocystis pneumonia with temozolomide chemoradiotherapy. J Neurooncol 2022; 159:665-674. [PMID: 35932358 DOI: 10.1007/s11060-022-04109-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/28/2022] [Indexed: 11/24/2022]
Abstract
INTRODUCTION The treatment of glioma with temozolomide chemoradiotherapy predisposes patients to pneumocystis pneumonia (PCP). Because PCP is a rare outcome, very little is known about specific clinical risk factors for its development in patients with glioma. METHODS We performed a population-based retrospective cohort study of glioma patients undergoing temozolomide chemoradiotherapy 2005 to 2019 in Ontario, Canada. We compared clinical features of patients who did not versus did develop PCP within one year of chemoradiotherapy. We examined the overall survival of patients by PCP status. RESULTS There were 5130 patients with glioma treated with temozolomide chemoradiotherapy. Ultimately, 38 patients (0.74%) were diagnosed with PCP within 1 year of chemoradiotherapy. Most (71%) infections occurred between 0-90 days and 29% occurred between 91-365 days. Median survival was 12.3 months in patients who did not develop PCP and 8.6 months in those who did develop PCP (P < 0.001). Trough 90-day lymphocyte counts were lower in the PCP group. When the lymphocytes fell below 0.19 × 109/L (or 0.25 × 109/L among patients without PCP prophylaxis), the risk of PCP was > 3.5%. CONCLUSIONS Pneumocystis pneumonia is rare in glioma patients who receive temozolomide chemoradiotherapy. Infection is associated with shorter survival and the development of lymphopenia. Reserving PCP prophylaxis for patients whose lymphocyte counts drop below 0.25 × 109/L may be a reasonable strategy.
Collapse
Affiliation(s)
- Seth A Climans
- Department of Oncology, Western University, London, Canada. .,Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Canada. .,London Regional Cancer Program, 800 Commissioners Rd E, London, ON, N6A5W9, Canada.
| | - Warren P Mason
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada
| | - Eva Grunfeld
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Canada
| | - Kelvin Chan
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Canada
| |
Collapse
|
18
|
Abstract
Given its aggressive natural history and immunosuppressive nature, glioblastoma (GBM) remains difficult to treat. Tumor Treating Fields (TTFields) are a promising treatment for GBM patients, yet the entirety of their antitumor action has not been fully elucidated. In a recent issue of the JCI, Chen et al. explored the effect of TTFields in reinvigorating immune responses. By elegant step-by-step approaches, the authors demonstrated that TTFields promote the production of immune-stimulating proinflammatory and interferon type 1 cytokines in tumor cells in a cGAS/STING- and AIM2 inflammasome–dependent mechanism, thereby activating the immune system. The findings show that TTFields not only directly inhibit tumor cell growth, as previously reported, but enhance antitumor immunity, suggesting TTFields can be used as an immune-modulating approach in GBM.
Collapse
Affiliation(s)
- Juyeun Lee
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute and
| | - Matthew M. Grabowski
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute and
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Cleveland, Ohio, USA
| | - Justin D. Lathia
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute and
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Cleveland, Ohio, USA
| |
Collapse
|
19
|
Candiota AP, Arús C. Establishing Imaging Biomarkers of Host Immune System Efficacy during Glioblastoma Therapy Response: Challenges, Obstacles and Future Perspectives. Metabolites 2022; 12:metabo12030243. [PMID: 35323686 PMCID: PMC8950145 DOI: 10.3390/metabo12030243] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/04/2022] [Accepted: 03/10/2022] [Indexed: 11/16/2022] Open
Abstract
This hypothesis proposal addresses three major questions: (1) Why do we need imaging biomarkers for assessing the efficacy of immune system participation in glioblastoma therapy response? (2) Why are they not available yet? and (3) How can we produce them? We summarize the literature data supporting the claim that the immune system is behind the efficacy of most successful glioblastoma therapies but, unfortunately, there are no current short-term imaging biomarkers of its activity. We also discuss how using an immunocompetent murine model of glioblastoma, allowing the cure of mice and the generation of immune memory, provides a suitable framework for glioblastoma therapy response biomarker studies. Both magnetic resonance imaging and magnetic resonance-based metabolomic data (i.e., magnetic resonance spectroscopic imaging) can provide non-invasive assessments of such a system. A predictor based in nosological images, generated from magnetic resonance spectroscopic imaging analyses and their oscillatory patterns, should be translational to clinics. We also review hurdles that may explain why such an oscillatory biomarker was not reported in previous imaging glioblastoma work. Single shot explorations that neglect short-term oscillatory behavior derived from immune system attack on tumors may mislead actual response extent detection. Finally, we consider improvements required to properly predict immune system-mediated early response (1–2 weeks) to therapy. The sensible use of improved biomarkers may enable translatable evidence-based therapeutic protocols, with the possibility of extending preclinical results to human patients.
Collapse
Affiliation(s)
- Ana Paula Candiota
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Cerdanyola del Vallès, 08193 Barcelona, Spain;
- Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Biociències, Edifici Cs, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain
| | - Carles Arús
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Cerdanyola del Vallès, 08193 Barcelona, Spain;
- Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Biociències, Edifici Cs, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain
- Correspondence:
| |
Collapse
|
20
|
da Silveira LM, Pedra NS, Bona NP, Spohr L, da Silva Dos Santos F, Saraiva JT, Alvez FL, de Moraes Meine B, Spanevello RM, Stefanello FM, Soares MSP. Selective in vitro anticancer effect of blueberry extract (Vaccinium virgatum) against C6 rat glioma: exploring their redox status. Metab Brain Dis 2022; 37:439-449. [PMID: 34748129 DOI: 10.1007/s11011-021-00867-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 10/31/2021] [Indexed: 10/19/2022]
Abstract
The aim of this study was to investigate the anticancer potential of blueberry extract (Vaccinium virgatum) against a C6 rat glioma lineage. Cultures of the C6 cells were exposed to blueberry extract at concentrations of 50 to 600 µg/mL for 12, 24, 48, or 72 h and then evaluated for cell viability, proliferation, migration, colony formation and oxidative stress. We also evaluated the effects of blueberry extract on primary rat cortical astrocytes. Our results show that treatment with blueberry extract did not alter the viability or proliferation of normal primary astrocytes but it did significantly reduce the viability in 21.54 % after 48 h and proliferation in 8.59 % after 24 h of C6 cells at 200 µg/mL. We also observed a reduction in the size of the colonies of 29.99 % at 100 µg/mL when compared to the control cells and cell migration was also reduced at 50 µg/mL. After 72 h, there was a reduction in the reactive oxygen species levels ranging from 46.26 to 34.73 %, in addition to a 380.2 % increase in total thiol content. Superoxide dismutase, catalase, and glutathione S-transferase activities were also enhanced when compared to the control. Taken together this data suggests that blueberry extract exerts some selective anticancer activity in C6 glioma cells.
Collapse
Affiliation(s)
- Larissa Menezes da Silveira
- Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, CEP: 96010-900, Brazil
| | - Nathalia Stark Pedra
- Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, CEP: 96010-900, Brazil
| | - Natália Pontes Bona
- Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Luiza Spohr
- Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, CEP: 96010-900, Brazil
| | - Francieli da Silva Dos Santos
- Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Juliane Torchelsen Saraiva
- Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Fernando Lopez Alvez
- Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, CEP: 96010-900, Brazil
| | - Bernardo de Moraes Meine
- Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Roselia Maria Spanevello
- Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, CEP: 96010-900, Brazil
| | - Francieli Moro Stefanello
- Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Mayara Sandrielly Pereira Soares
- Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, CEP: 96010-900, Brazil.
| |
Collapse
|
21
|
Zhang Y, Chen S, Chen H, Chen S, Li Z, Feng E, Li W. Prognostic Value and Risk Factors of Treatment-Related Lymphopenia in Malignant Glioma Patients Treated With Chemoradiotherapy: A Systematic Review and Meta-Analysis. Front Neurol 2022; 12:726561. [PMID: 35058869 PMCID: PMC8764122 DOI: 10.3389/fneur.2021.726561] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 12/09/2021] [Indexed: 01/11/2023] Open
Abstract
Background: Immunotherapy has shown promising therapeutic efficacy in various cancers but not gliomas. Circulating lymphocytes play critical roles in cancer control and responses to immune checkpoint inhibitors. Treatment-related lymphopenia has been associated with poor survival in patients with various tumors. This meta-analysis evaluated the risk and impact of lymphopenia in patients with glioma. Methods: The PubMed, Embase, Web of Science, and Cochrane Library databases were comprehensively searched. Eligible studies were included if they reported the incidence and risk factors of lymphopenia and the impact of lymphopenia on survival. Stata 16.0 was used for this meta-analysis. Results: A total of 21 studies were included in the final systematic review and 20 were included in the quantitative analysis. The overall incidence of grade III/IV lymphopenia was 31.6% [95% confidence interval (CI), 22.3-40.8%]. Pooled results based on pathology of glioma revealed that the incidence in astrocytoma and astrocytoma oligodendroglioma patients was 20.2% (95% CI:5.9-34.4%), and the incidence in glioblastoma patients was 27.6% (95% CI:16.2-38.9%). Lymphopenia was associated with poor overall survival (hazard ratio, 1.99; 95% CI, 1.74-2.27; P< 0.001) compared to no lymphopenia. Brain receiving radiation dose of 20 or 25 Gy, female sex, older age, lower baseline lymphocyte count, and dexamethasone dose > 2 mg instead of baseline use were risk factors for lymphopenia. Conclusions: Treatment-related lymphopenia was associated with decreased survival in patients with glioma. Optimization of chemoradiation regimens, particularly in patients with concurrent risk factors, can reduce lymphopenia and potentially improve survival in the era of immunotherapy.
Collapse
Affiliation(s)
- Yongchao Zhang
- Cancer Center, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Shichao Chen
- Neurosurgery Department, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Hualei Chen
- Cancer Center, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Shanshan Chen
- Cancer Center, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Zhen Li
- Emergency Department, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Enshan Feng
- Neurosurgery Department, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Wei Li
- Cancer Center, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
22
|
Arnesen VS, Gras Navarro A, Chekenya M. Challenges and Prospects for Designer T and NK Cells in Glioblastoma Immunotherapy. Cancers (Basel) 2021; 13:4986. [PMID: 34638471 PMCID: PMC8507952 DOI: 10.3390/cancers13194986] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 12/22/2022] Open
Abstract
Glioblastoma (GBM) is the most prevalent, aggressive primary brain tumour with a dismal prognosis. Treatment at diagnosis has limited efficacy and there is no standardised treatment at recurrence. New, personalised treatment options are under investigation, although challenges persist for heterogenous tumours such as GBM. Gene editing technologies are a game changer, enabling design of novel molecular-immunological treatments to be used in combination with chemoradiation, to achieve long lasting survival benefits for patients. Here, we review the literature on how cutting-edge molecular gene editing technologies can be applied to known and emerging tumour-associated antigens to enhance chimeric antigen receptor T and NK cell therapies for GBM. A tight balance of limiting neurotoxicity, avoiding tumour antigen loss and therapy resistance, while simultaneously promoting long-term persistence of the adoptively transferred cells must be maintained to significantly improve patient survival. We discuss the opportunities and challenges posed by the brain contexture to the administration of the treatments and achieving sustained clinical responses.
Collapse
Affiliation(s)
| | - Andrea Gras Navarro
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, 5009 Bergen, Norway
| | - Martha Chekenya
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, 5009 Bergen, Norway
| |
Collapse
|
23
|
Mistry AM, Jonathan SV, Monsour MA, Mobley BC, Clark SW, Moots PL. Impact of postoperative dexamethasone on survival, steroid dependency, and infections in newly diagnosed glioblastoma patients. Neurooncol Pract 2021; 8:589-600. [PMID: 34594571 PMCID: PMC8475235 DOI: 10.1093/nop/npab039] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND We examined the effect of dexamethasone prescribed in the initial 3 postoperative weeks on survival, steroid dependency, and infection in glioblastoma patients. METHODS In this single-center retrospective cohort analysis, we electronically retrieved inpatient administration and outpatient prescriptions of dexamethasone and laboratory values from the medical record of 360 glioblastoma patients. We correlated total dexamethasone prescribed from postoperative day (POD) 0 to 21 with survival, dexamethasone prescription from POD30 to POD90, and diagnosis of an infection by POD90. These analyses were adjusted for age, Karnofsky performance status score, tumor volume, extent of resection, IDH1/2 tumor mutation, tumor MGMT promoter methylation, temozolomide and radiotherapy initiation, and maximum blood glucose level. RESULTS Patients were prescribed a median of 159 mg [109-190] of dexamethasone cumulatively by POD21. Every 16-mg increment (4 mg every 6 hours/day) of total dexamethasone associated with a 4% increase in mortality (95% confidence interval [CI] 1%-7%, P < .01), 12% increase in the odds of being prescribed dexamethasone from POD30 to POD90 (95% CI 6%-19%, P < .01), and 10% increase in the odds of being diagnosed with an infection (95% CI, 4%-17%, P < .01). Of the 175 patients who had their absolute lymphocyte count measured in the preoperative week, 80 (45.7%) had a value indicative of lymphopenia. In the POD1-POD28 period, this proportion was 82/167 (49.1%). CONCLUSIONS Lower survival, steroid dependency, and higher infection rate in glioblastoma patients associated with higher dexamethasone administration in the initial 3 postoperative weeks. Nearly half of the glioblastoma patients are lymphopenic preoperatively and up to 1 month postoperatively.
Collapse
Affiliation(s)
- Akshitkumar M Mistry
- Department of Neurological Surgery, University of Louisville, Louisville, Kentucky, USA
| | | | | | - Bret C Mobley
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Stephen W Clark
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Paul L Moots
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
24
|
Brault C, Zerbib Y, Chouaki T, Maizel J, Nyga R. Temozolomide is a risk factor for invasive pulmonary aspergillosis: A case report and literature review. Infect Dis Now 2021; 51:630-632. [PMID: 34581280 DOI: 10.1016/j.idnow.2020.11.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 11/12/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Temozolomide is an oral alkylating agent incorporated in the treatment of glioblastoma multiforme (GBM) that can lead to lymphopenia. The standard treatment of GBM involves temozolomide chemotherapy with radiation, often with addition of corticosteroids for symptomatic management of cerebral edema. Some studies have reported an increased risk of opportunistic infections. CASE PRESENTATION A 72-year-old man receiving Temozolomide for treatment of newly diagnosed GBM associated with radiotherapy and corticosteroids was admitted in an intensive care unit because a rapid deterioration of consciousness associated with acute respiratory failure. The diagnosis of invasive pulmonary aspergillosis (IPA) was made. The patient was successfully treated with voriconazole alone. CONCLUSIONS This case shows that Temozolomide can be associated with severe invasive aspergillosis, which is in all likelihood associated with T lymphocyte immune dysfunction. Physicians should be aware of possible opportunistic infections when managing patients with glioblastoma, and patients exposed to this agent should be carefully monitored.
Collapse
Affiliation(s)
- Clément Brault
- Department of Intensive Care Medicine, CHU Amiens-Picardie, avenue Laennec, 80000 Amiens, France.
| | - Yoann Zerbib
- Department of Intensive Care Medicine, CHU Amiens-Picardie, avenue Laennec, 80000 Amiens, France
| | - Taieb Chouaki
- Parasitology and Mycology Department, Amiens University Hospital, 80000 Amiens, France
| | - Julien Maizel
- Department of Intensive Care Medicine, CHU Amiens-Picardie, avenue Laennec, 80000 Amiens, France
| | - Rémy Nyga
- Department of Intensive Care Medicine, CHU Amiens-Picardie, avenue Laennec, 80000 Amiens, France
| |
Collapse
|
25
|
Zhou J, Chen W, Liu Y, Qu C, Jiang W, Yin J, Lin J, Mao W, Ye B, Zhou J, Ke L, Tong Z, Liu Y, Li W. Trajectories of Lymphocyte Counts in the Early Phase of Acute Pancreatitis Are Associated With Infected Pancreatic Necrosis. Clin Transl Gastroenterol 2021; 12:e00405. [PMID: 34597275 PMCID: PMC8462575 DOI: 10.14309/ctg.0000000000000405] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 08/17/2021] [Indexed: 01/09/2023] Open
Abstract
INTRODUCTION Infected pancreatic necrosis (IPN) is an important complication of acute pancreatitis (AP). Absolute lymphocyte count (ALC) was reported to be associated with immunosuppression and the development of IPN. The aim of this study was to describe the trajectory of ALC during the early phase of AP and assess its association with IPN. METHODS We retrospectively screened patients with AP admitted to our center between January 2016 and July 2019. The ALC levels for the first 7 days after admission were collected. Group-based trajectory modeling was performed to detect the trajectories. Cox proportional hazards regression model was adopted to identify potential risk factors of IPN. RESULTS Overall, 292 patients were enrolled for analysis. A triple-group trajectory model was developed, assigning 116 patients to the low-level ALC group, 133 to the medium-level ALC group, and 43 to the high-level ALC group. There was no overall significant difference regarding the incidence of IPN among the 3 groups (P = 0.066). In pairwise comparison, patients in the low-level ALC group had significantly higher incidence of IPN than those in the high-level ALC group (hazard ratio: 3.50; 95% confidence interval: 1.22-10.00, P = 0.020). Length of hospital stay and intensive care unit stay differed significantly among patients with different trajectories (P = 0.042 and 0.033, respectively). DISCUSSION Despite the fact that the trajectories of ALC is overall insignificant for the development of IPN, patients with persistent low ALC trajectories during the early phase of AP are more likely to develop IPN when compared with patients with high ALC trajectories.
Collapse
Affiliation(s)
- Jing Zhou
- Department of Critical Care Medicine, Jinling Hospital, Medical School of Southeast University, Nanjing, China;
| | - Wensong Chen
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China;
| | - Yang Liu
- Department of Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Cheng Qu
- Department of Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wendi Jiang
- Department of Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jiangtao Yin
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China;
| | - Jiajia Lin
- Department of Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wenjian Mao
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China;
- Department of Biostatistics, School of Public Health, Southern Medical University, Guangzhou, China.
| | - Bo Ye
- Department of Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jing Zhou
- Department of Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lu Ke
- Department of Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
- National Institute of Healthcare Data Science at Nanjing University
| | - Zhihui Tong
- Department of Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yuxiu Liu
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China;
- Department of Biostatistics, School of Public Health, Southern Medical University, Guangzhou, China.
| | - Weiqin Li
- Department of Critical Care Medicine, Jinling Hospital, Medical School of Southeast University, Nanjing, China;
- Department of Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
- National Institute of Healthcare Data Science at Nanjing University
| |
Collapse
|
26
|
Asano K, Fumoto T, Matsuzaka M, Hasegawa S, Suzuki N, Akasaka K, Katayama K, Kamataki A, Kurose A, Ohkuma H. Combination chemoradiotherapy with temozolomide, vincristine, and interferon-β might improve outcomes regardless of O6-methyl-guanine-DNA-methyltransferase (MGMT) promoter methylation status in newly glioblastoma. BMC Cancer 2021; 21:867. [PMID: 34320929 PMCID: PMC8320052 DOI: 10.1186/s12885-021-08592-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 07/05/2021] [Indexed: 12/30/2022] Open
Abstract
Background This investigator-initiated, open-label, single-arm, single-institute study was conducted to investigate the effectiveness of induction combination chemoradiotherapy and long-term maintenance therapy with temozolomide (TMZ) plus interferon (IFN)-β for glioblastoma. Methods The initial induction combination chemoradiotherapy comprised radiotherapy plus TMZ plus vincristine plus IFN-β. Maintenance chemotherapy comprised monthly TMZ, continued for 24–50 cycles, plus weekly IFN-β continued for as long as possible. The primary endpoint was 2-year overall survival (2y-OS). The study protocol was to be considered valid if the expected 2y-OS was over 38% and the lower limit of the 95% confidence interval (CI) was no less than 31.7% compared with historical controls, using Kaplan-Meier methods. Secondary endpoints were median progression-free survival (mPFS), median OS (mOS), 5-year OS rate (5y-OS), and mPFS and mOS classified according to MGMT promoter methylation status. Results Forty-seven patients were analyzed. The 2y-OS was 40.7% (95%CI, 27.5–55.4%). The mPFS and mOS were 11.0 months and 18.0 months, respectively, and 5y-OS was 20.3% (95%CI, 10.9–34.6%). The mPFS in groups with and without MGMT promoter methylation in the tumor was 10.0 months and 11.0 months (p = 0.59), respectively, and mOS was 24.0 months and 18.0 months (p = 0.88), respectively. The frequency of grade 3/4 neutropenia was 19.1%. Conclusions The 2y-OS with induction multidrug combination chemoradiotherapy and long-term maintenance therapy comprising TMZ plus IFN-β tended to exceed that of historical controls, but the lower limit of the 95%CI was below 31.7%. Although the number of cases was small, this protocol may rule out MGMT promoter methylation status as a prognostic factor. Trial registration University Hospital Medical Information Network (number UMIN000040599).
Collapse
Affiliation(s)
- Kenichiro Asano
- Department of Neurosurgery, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan.
| | - Toshio Fumoto
- Department of Neurosurgery, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
| | - Masashi Matsuzaka
- Clinical Research Support Center, Hirosaki University Hospital, 53 Hon-cho, Hirosaki, Aomori, 036-8563, Japan.,Department of Medical Informatics, Hirosaki University Hospital, 53 Hon-cho, Hirosaki, Aomori, 036-8563, Japan
| | - Seiko Hasegawa
- Department of Neurosurgery, Kuroishi General Hospital, 1-70 Kitami-cho, Kuroishi, Aomori, 036-0541, Japan
| | - Naoya Suzuki
- Department of Neurosurgery, Towada City Hospital, 8-14 Nishi-Jyuniban-cho, Towada, Aomori, 034-0093, Japan
| | - Kenichi Akasaka
- Department of Neurosurgery, Towada City Hospital, 8-14 Nishi-Jyuniban-cho, Towada, Aomori, 034-0093, Japan
| | - Kosuke Katayama
- Department of Neurosurgery, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
| | - Akihisa Kamataki
- Department of Anatomic Pathology, Hirosaki University Graduate School of Medicine, 53 Honcho, Hirosaki, Aomori, 036-8563, Japan
| | - Akira Kurose
- Department of Anatomic Pathology, Hirosaki University Graduate School of Medicine, 53 Honcho, Hirosaki, Aomori, 036-8563, Japan
| | - Hiroki Ohkuma
- Department of Neurosurgery, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
| |
Collapse
|
27
|
Gonakoti S, Parra-Rodriguez L. Should CD4 Levels be Monitored in a Patient on Temozolomide? Eur J Case Rep Intern Med 2021; 8:002276. [PMID: 33987112 DOI: 10.12890/2021_002276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 02/25/2021] [Indexed: 11/05/2022] Open
Abstract
Introduction Temozolomide is an alkylating agent, indicated in the treatment of refractory anaplastic astrocytoma and newly diagnosed glioblastoma. We describe a case of Salmonella typhimurium bacteraemia associated with septic arthritis in the setting of concurrent temozolomide use. Case A 67-year-old woman presented with acute onset of bilateral knee pain and swelling. She had a medical history of glioblastoma multiforme treated with temozolomide. Synovial fluid analysis and blood cultures revealed S. typhimurium, confirming a diagnosis of S. typhimurium bacteraemia associated with septic arthritis. Conclusion We conclude that chemotherapy with temozolomide and corticosteroid use will increase an individual's susceptibility to a wide variety of opportunistic infections akin to HIV-associated acquired immunodeficiency syndrome (AIDS). Furthermore, we hypothesize a possible benefit of monitoring CD4 levels and prophylaxis against opportunistic infections (based on the CD4 levels) in individuals receiving temozolomide-based chemotherapy, similar to HIV-AIDS. LEARNING POINTS Chemotherapy with temozolomide and corticosteroid use will increase an individual's susceptibility to a wide variety of opportunistic infections akin to HIV-associated acquired immunodeficiency syndrome (AIDS).There may be a benefit in monitoring CD4 levels in patients who are receiving temozolomide.Given selective CD4 lymphopenia while on temozolomide, there may be a possible benefit of prophylaxis against opportunistic infections in individuals based on their CD4 levels, similar to HIV-AIDS (current guidelines recommend considering Pneumocystis jirovecii pneumonia prophylaxis for patients receiving concomitant radiotherapy and temozolomide, and high-dose steroids).
Collapse
Affiliation(s)
- Sriram Gonakoti
- John H Stroger Jr Hospital of Cook County, Chicago, Illinois, USA
| | | |
Collapse
|
28
|
Ortiz R, Perazzoli G, Cabeza L, Jiménez-Luna C, Luque R, Prados J, Melguizo C. Temozolomide: An Updated Overview of Resistance Mechanisms, Nanotechnology Advances and Clinical Applications. Curr Neuropharmacol 2021; 19:513-537. [PMID: 32589560 PMCID: PMC8206461 DOI: 10.2174/1570159x18666200626204005] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 06/17/2020] [Accepted: 06/21/2020] [Indexed: 12/22/2022] Open
Abstract
Temozolomide (TMZ), an oral alkylating prodrug which delivers a methyl group to purine bases of DNA (O6-guanine; N7-guanine and N3-adenine), is frequently used together with radiotherapy as part of the first-line treatment of high-grade gliomas. The main advantages are its high oral bioavailability (almost 100% although the concentration found in the cerebrospinal fluid was approximately 20% of the plasma concentration of TMZ), its lipophilic properties, and small size that confer the ability to cross the blood-brain barrier. Furthermore, this agent has demonstrated activity not only in brain tumors but also in a variety of solid tumors. However, conventional therapy using surgery, radiation, and TMZ in glioblastoma results in a median patient survival of 14.6 months. Treatment failure has been associated with tumor drug resistance. This phenomenon has been linked to the expression of O6-methylguanine-DNA methyltransferase, but the mismatch repair system and the presence of cancer stem-like cells in tumors have also been related to TMZ resistance. The understanding of these mechanisms is essential for the development of new therapeutic strategies in the clinical use of TMZ, including the use of nanomaterial delivery systems and the association with other chemotherapy agents. The aim of this review is to summarize the resistance mechanisms of TMZ and the current advances to improve its clinical use.
Collapse
Affiliation(s)
- Raúl Ortiz
- Institute of Biopathology and Regenerative Medicine (IBIMER), Biomedical Research Centre (CIBM), University of Granada, Spain
| | | | - Laura Cabeza
- Institute of Biopathology and Regenerative Medicine (IBIMER), Biomedical Research Centre (CIBM), University of Granada, Spain
| | - Cristina Jiménez-Luna
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Epalinges 1066, Switzerland
| | - Raquel Luque
- Medical Oncology Service, Virgen de las Nieves Hospital, Granada, Spain
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Biomedical Research Centre (CIBM), University of Granada, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Biomedical Research Centre (CIBM), University of Granada, Spain
| |
Collapse
|
29
|
Temozolomide treatment outcomes and immunotherapy efficacy in brain tumor. J Neurooncol 2021; 151:55-62. [PMID: 32813186 PMCID: PMC9833842 DOI: 10.1007/s11060-020-03598-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 08/08/2020] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Glioblastoma (GBM) has a survival rate of around 2 years with aggressive current standard of care. While other tumors have responded favorably to trials combining immunotherapy and chemotherapy, GBM remains uniformly deadly with minimal increases in overall survival. GBM differ from others due to being isolated behind the blood brain barrier, increased heterogeneity and mutational burden, and immunosuppression from the brain environment and tumor itself. METHODS We have reviewed clinical and preclinical studies investigating how different doses (dose intense (DI) and metronomic) and timing of immunotherapy following TMZ treatment can eradicate tumor cells, alter tumor mutational burden, and change immune cells. RESULTS Recent clinical trials with standard of care (SoC), DI and metronomic TMZ regimes are no able to completely eradicate GBM. Elevated TMZ levels in DI treatment can overcome MGMT resistance but may result in hypermutation of surviving tumor cells. Higher levels of TMZ will also generate a higher degree of lymphopenia compared to SoC and metronomic regimes in preclinical studies. CONCLUSION The different levels of lymphopenia and tumor eradication discussed in this review suggest possible beneficial pairings between immunotherapy and TMZ treatment. Treatments resulting in profound lymphopenia will allow for expansion of vaccine specific T cells or of CAT T cells. Clinical and preclinical studies are currently comparing different combinations of TMZ and immunotherapy timing to treat GBM through a balance between tumor killing and immune cell expansion. More frequent immune monitoring time points in ongoing clinical trials are crucial for further development of these combinations.
Collapse
|
30
|
Exceptional responses with sequential metronomic temozolomide after pembrolizumab failure in patients with metastatic melanoma. Melanoma Res 2020; 29:643-647. [PMID: 30829928 DOI: 10.1097/cmr.0000000000000592] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Pembrolizumab is an effective therapy for patients with metastatic melanoma. However, not all patients derive benefit. It is postulated that an increase in regulatory T cells in melanoma patients can impair the response to immunotherapies. Continuous low-dose temozolomide has shown to cause immunomodulatory effects resulting in CD4 + lymphopenia due to which Treg population can also decrease significantly. Herein, we present a case series of three patients with metastatic melanoma who after progression on pembrolizumab showed a radiological response after just one cycle of metronomic temozolomide (75 mg/m daily for 6 weeks on 8-week cycle). This suggests that temozolomide may be a useful alternative for patients with metastatic melanoma after disease progression on pembrolizumab. Further studies with biomarkers are warranted to elucidate which patients will derive benefit from this strategy.
Collapse
|
31
|
Lee HN, Koo HJ, Kim SH, Choi SH, Sung H, Do KH. Human Bocavirus Infection in Adults: Clinical Features and Radiological Findings. Korean J Radiol 2020; 20:1226-1235. [PMID: 31270986 PMCID: PMC6609429 DOI: 10.3348/kjr.2018.0634] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Accepted: 04/07/2019] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE Human bocavirus (HBoV) is a newly identified pathogen that can cause upper and lower respiratory infections usually in children; however, its clinical characteristics and significance in respiratory infections in adults have not been well known. Our objective was to evaluate the clinical features of respiratory HBoV infection and to describe the CT findings of HBoV pneumonia in adults. MATERIALS AND METHODS A total of 185 adult patients diagnosed with HBoV infection at a tertiary referral center between January 2010 and December 2017 were retrospectively evaluated with respect to the clinical characteristics of HBoV infection and its risk factors for pneumonia. Chest CT findings for 34 patients with HBoV pneumonia without co-infection were analyzed and compared between immunocompetent (n = 18) and immunocompromised (n = 16) patients. RESULTS HBoV infections were predominantly noted between February and June. Among the 185 patients with HBoV infection, 119 (64.3%) had community-acquired infections and 110 (59.5%) had pneumonia. In multivariable analysis, older age (odds ratio [OR], 1.02; 95% confidence interval [CI], 1.00-1.04; p = 0.045) and nosocomial infection (OR, 2.07; 95% CI, 1.05-4.10; p = 0.037) were associated with HBoV pneumonia. The main CT findings were bilateral consolidation (70.6%) and/or ground-glass opacities (64.7%); centrilobular nodules (14.7%) were found less frequently. The pattern of CT findings were not significantly different between immunocompetent and immunocompromised patients (all, p > 0.05). CONCLUSION HBoV infection can be a potential respiratory tract infection in adults. The most frequent CT findings of HBoV pneumonia were bilateral consolidation and/or ground-glass opacities.
Collapse
Affiliation(s)
- Han Na Lee
- Department of Radiology, Kyung Hee University Hospital at Gangdong, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Hyun Jung Koo
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Soo Hyun Kim
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sang Ho Choi
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Heungsup Sung
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyung Hyun Do
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
32
|
Kleinberg L, Sloan L, Grossman S, Lim M. Radiotherapy, Lymphopenia, and Host Immune Capacity in Glioblastoma: A Potentially Actionable Toxicity Associated With Reduced Efficacy of Radiotherapy. Neurosurgery 2020; 85:441-453. [PMID: 31232425 DOI: 10.1093/neuros/nyz198] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 02/24/2019] [Indexed: 12/14/2022] Open
Abstract
Radiotherapy is cytotoxic to tumor cells and is therefore a critical component of therapy for many malignancies, including glioblastoma (GBM). We now appreciate the value of the immunomodulatory effects of radiation that may be important to overall therapeutic success in some patients with this primary brain tumor. Although potentially beneficial immune-stimulating properties of radiotherapy treatment have been the focus of recent study, this modality is actually at the same time associated with the depletion of lymphocytes, which are crucial to the defense against neoplastic development and progression. In this review, we describe the association of systemic lymphopenia with poor tumor outcome, present evidence that radiotherapy is an important contributing cause of lymphodepletion, describe the systemic immune context of tumor and brain injury that contributes to immunosuppression, describe other contributing factors to lymphopenia including concomitant medications and treatments, and speculate about the role of the normal physiologic response to brain injury in the immunosuppressive dynamics of GBM. Radiotherapy is one significant and potentially actionable iatrogenic suppressor of immune response that may be limiting the success of therapy in GBM and other tumor types. Altered strategies for radiotherapy more permissive of a vigorous antineoplastic immune response may improve outcome for malignancy.
Collapse
Affiliation(s)
- Lawrence Kleinberg
- Department of Radiation Oncology and Radiation Molecular Sciences, Johns Hopkins University, Baltimore, Maryland
| | - Lindsey Sloan
- Department of Radiation Oncology and Radiation Molecular Sciences, Johns Hopkins University, Baltimore, Maryland
| | - Stuart Grossman
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland
| | - Michael Lim
- Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
33
|
Abstract
Gliomas, the most common malignant primary brain tumours, remain universally lethal. Yet, seminal discoveries in the past 5 years have clarified the anatomy, genetics and function of the immune system within the central nervous system (CNS) and altered the paradigm for successful immunotherapy. The impact of standard therapies on the response to immunotherapy is now better understood, as well. This new knowledge has implications for a broad range of tumours that develop within the CNS. Nevertheless, the requirements for successful therapy remain effective delivery and target specificity, while the dramatic heterogeneity of malignant gliomas at the genetic and immunological levels remains a profound challenge.
Collapse
Affiliation(s)
- John H Sampson
- The Preston Robert Tisch Brain Tumor Center at Duke, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA.
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA.
| | - Michael D Gunn
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Peter E Fecci
- The Preston Robert Tisch Brain Tumor Center at Duke, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
- Duke Center for Brain and Spine Metastasis, Duke University Medical Center, Durham, NC, USA
| | - David M Ashley
- The Preston Robert Tisch Brain Tumor Center at Duke, Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
34
|
Karachi A, Dastmalchi F, Mitchell DA, Rahman M. Temozolomide for immunomodulation in the treatment of glioblastoma. Neuro Oncol 2019; 20:1566-1572. [PMID: 29733389 DOI: 10.1093/neuonc/noy072] [Citation(s) in RCA: 185] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Temozolomide is the most widely used chemotherapy for patients with glioblastoma (GBM) despite the fact that approximately half of treated patients have temozolomide resistance and all patients eventually fail therapy. Due to the limited efficacy of existing therapies, immunotherapy is being widely investigated for patients with GBM. However, initial immunotherapy trials in GBM patients have had disappointing results as monotherapy. Therefore, combinatorial treatment strategies are being investigated. Temozolomide has several effects on the immune system that are dependent on mode of delivery and the dosing strategy, which may have unpredicted effects on immunotherapy. Here we summarize the immune modulating role of temozolomide alone and in combination with immunotherapies such as dendritic cell vaccines, T-cell therapy, and immune checkpoint inhibitors for patients with GBM.
Collapse
Affiliation(s)
- Aida Karachi
- Lillian S. Wells Department of Neurosurgery, UF Brain Tumor Immunotherapy Program, University of Florida, Gainesville, Florida
| | - Farhad Dastmalchi
- Lillian S. Wells Department of Neurosurgery, UF Brain Tumor Immunotherapy Program, University of Florida, Gainesville, Florida
| | - Duane A Mitchell
- Lillian S. Wells Department of Neurosurgery, UF Brain Tumor Immunotherapy Program, University of Florida, Gainesville, Florida
| | - Maryam Rahman
- Lillian S. Wells Department of Neurosurgery, UF Brain Tumor Immunotherapy Program, University of Florida, Gainesville, Florida
| |
Collapse
|
35
|
Liu SA, Sullivan T, Bryce C, Chan AM, Cilmi S. Cerebral aspergillosis within new tumour site presents as incidental new brain lesion in patient receiving temozolomide for glioblastoma multiforme. BMJ Case Rep 2019; 12:12/5/e227500. [PMID: 31154345 DOI: 10.1136/bcr-2018-227500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Glioblastoma multiforme (GBM) is an aggressive tumour that can lead to lymphopaenia. Its standard treatment involves temozolomide (TMZ) chemotherapy with radiation, often with addition of corticosteroids for symptomatic management. Although TMZ is also immunosuppressive, patients receiving TMZ rarely develop disseminated opportunistic infections. Here, we report the case of a patient with GBM receiving TMZ, radiotherapy and corticosteroids, who develops an incidental new brain lesion that is found to be disseminated Aspergillus within a new GBM tumour site. The patient received successful early treatment of her central nervous system aspergillosis. This case illustrates the profound immunosuppressive potential of GBM in conjunction with TMZ and corticosteroids, which can lead to high-morbidity opportunistic infections concurrently with tumour progression. Future research is needed to elucidate GBM, TMZ and corticosteroids' compound immune effects and guide management that strikes a balance between treating high-morbidity infections and continuing with immunosuppressive chemotherapy.
Collapse
Affiliation(s)
- Shiyuan Anabeth Liu
- Internal Medicine, Washington University in Saint Louis School of Medicine, Saint Louis, Missouri, USA
| | - Timothy Sullivan
- Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Clare Bryce
- Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Amy M Chan
- Internal Medicine, Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Salvatore Cilmi
- Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
36
|
Ye LL, Fan XW, Hu CS, He XY, Wang XS, Shen CY, Xu TT, Ying HM. Dosimetry of the brain and hypothalamus predicting acute lymphopenia and the survival of glioma patients with postoperative radiotherapy. Cancer Med 2019; 8:2759-2768. [PMID: 30983159 PMCID: PMC6558490 DOI: 10.1002/cam4.2159] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 03/29/2019] [Accepted: 03/29/2019] [Indexed: 01/20/2023] Open
Abstract
Background The aim of this study was to investigate dosimetric factors for predicting acute lymphopenia and the survival of glioma patients with postoperative intensity‐modulated radiotherapy (IMRT). Methods A total of 148 glioma patients were reviewed. Acute lymphopenia was defined as a peripheral lymphocyte count (PLC) lower than 1.0 × 109/L during radiotherapy with a normal level at pretreatment. PLCs with the corresponding dates and dose volume histogram parameters were collected. Univariate and multivariate Cox regression analyses were constructed to assess the significance of risk factors associated with lymphopenia and overall survival (OS). Results Sixty‐nine (46.6%) patients developed lymphopenia during radiotherapy. Multivariate analyses revealed that the risk increased with the maximal dose of the hypothalamus (HT Dmax) ≥56 Gy (58.9% vs 28.5%, P = 0.002), minimal dose of the whole brain (WB Dmin) ≥2 Gy (54.3% vs 33.9%, P = 0.006), or mean dose of the WB (WB Dmean) ≥34 Gy (56.0% vs 37.0%, P = 0.022). Patients with older age, high‐grade glioma, development of lymphopenia, high HT Dmax, WB Dmin, and WB Dmean had significantly inferior OS in the multivariate analyses. Conclusions HT Dmax, WB Dmin, and WB Dmean are promising indicators of lymphopenia and the survival of glioma patients undergoing postoperative IMRT. The necessity and feasibility of dosimetric constraints for HT and WB is warranted with further investigation.
Collapse
Affiliation(s)
- Lu-Lu Ye
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, P.R China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R China
| | - Xing-Wen Fan
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, P.R China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R China
| | - Chao-Su Hu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, P.R China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R China
| | - Xia-Yun He
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, P.R China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R China
| | - Xiao-Shen Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, P.R China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R China
| | - Chun-Ying Shen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, P.R China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R China
| | - Ting-Ting Xu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, P.R China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R China
| | - Hong-Mei Ying
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, P.R China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R China
| |
Collapse
|
37
|
Clinical observation of lymphopenia in patients with newly diagnosed glioblastoma. J Neurooncol 2019; 143:321-328. [DOI: 10.1007/s11060-019-03167-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 04/08/2019] [Indexed: 12/26/2022]
|
38
|
Furukawa R, Homma H, Inoue T, Horiuchi H, Usui K. Cytomegalovirus Hemorrhagic Cystitis in a Malignant Glioma Patient Treated with Temozolomide. Intern Med 2018; 57:3047-3050. [PMID: 29780148 PMCID: PMC6232021 DOI: 10.2169/internalmedicine.1005-18] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Temozolomide, a key drug in the treatment of malignant glioma, can cause profound lymphopenia and various opportunistic infectious diseases. A 79-year-old woman with anaplastic oligodendroglioma developed a fever and gross hematuria after 8 weeks of standard radiotherapy with concomitant temozolomide treatment. A cytomegalovirus (CMV) antigen test for pp65 antigenemia was positive (137 cells per 75,800 leukocytes), and the findings from a urine cytology test were consistent with CMV-induced hemorrhagic cystitis. She was treated with ganciclovir, and her condition improved. CMV monitoring is needed when patients develop symptoms related to opportunistic infections during temozolomide treatment for malignant glioma.
Collapse
Affiliation(s)
| | - Hirokuni Homma
- Department of Neurosurgery, NTT Medical Center Tokyo, Japan
| | - Tomohiro Inoue
- Department of Neurosurgery, NTT Medical Center Tokyo, Japan
| | - Hajime Horiuchi
- Department of Diagnostic Pathology, NTT Medical Center Tokyo, Japan
| | - Kazuhiro Usui
- Division of Respirology, NTT Medical Center Tokyo, Japan
| |
Collapse
|
39
|
KX2-361: a novel orally bioavailable small molecule dual Src/tubulin inhibitor that provides long term survival in a murine model of glioblastoma. J Neurooncol 2018; 140:519-527. [DOI: 10.1007/s11060-018-2992-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/27/2018] [Indexed: 10/28/2022]
|
40
|
Phytosomal curcumin causes natural killer cell-dependent repolarization of glioblastoma (GBM) tumor-associated microglia/macrophages and elimination of GBM and GBM stem cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:168. [PMID: 30041669 PMCID: PMC6058381 DOI: 10.1186/s13046-018-0792-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 06/14/2018] [Indexed: 11/10/2022]
Abstract
BACKGROUND Glioblastoma (GBM) is a primary brain tumor with a 5-year survival rate of ≤5%. We have shown earlier that GBM-antibody-linked curcumin (CC) and also phytosomal curcumin (CCP) rescue 50-60% of GBM-bearing mice while repolarizing the tumor-associated microglia/macrophages (TAM) from the tumor-promoting M2-type to the tumoricidal M1-type. However, systemic application of CCP yields only sub-IC50 concentrations of CC in the plasma, which is unlikely to kill GBM cells directly. This study investigates the role of CC-evoked intra-GBM recruitment of activated natural killer (NK) cells in the elimination of GBM and GBM stem cells. METHODS We have used an immune-competent syngeneic C57BL6 mouse model with the mouse-GBM GL261 cells orthotopically implanted in the brain. Using immunohistochemistry and flow cytometry, we have quantitatively analyzed the role of the intra-GBM-recruited NK cells by (i) injecting (i.p.) the NK1.1 antibody (NK1.1Ab) to temporarily eliminate the NK cells and (ii) blocking NK recruitment by injecting an IL12 antibody (IL12Ab). The treatment cohorts used randomly-chosen GL261-implanted mice and data sets were compared using two-tailed t-test or ANOVA. RESULTS CCP treatment caused the GBM tumor to acquire M1-type macrophages (50-60% of the TAM) and activated NK cells. The treatment also elicited (a) suppression of the M2-linked tumor-promoting proteins STAT3, ARG1, and IL10, (b) induction of the M1-linked anti-tumor proteins STAT1 and inducible nitric oxide synthase in the TAM, (c) elimination of CD133(+) GBM stem cells, and (d) activation of caspase3 in the GBM cells. Eliminating intra-GBM NK cell recruitment caused a partial reversal of each of these effects. Concomitantly, we observed a CCP-evoked dramatic induction of the chemokine monocyte chemotactic protein-1 (MCP-1) in the TAM. CONCLUSIONS The recruited NK cells mediate a major part of the CCP-evoked elimination of GBM and GBM stem cells and stabilization of the TAM in the M1-like state. MCP-1 is known to activate peripheral M1-type macrophages to secrete IL12, an activator of NK cells. Based on such observations, we postulate that by binding to peripheral M1-type macrophages and IL12-activated NK cells, the brain-released chemokine MCP-1 causes recruitment of peripheral immune cells into the GBM, thereby causing destruction of the GBM cells and GBM stem cells.
Collapse
|
41
|
Kao FC, Hsu YC, Lai PY, Wang CB, Tu YK, Chen WK. One-year mortality and Periprosthetic infection rates after Total knee Arthroplasty in Cancer patients: a population-based cohort study. BMC Cancer 2018; 18:628. [PMID: 29866058 PMCID: PMC5987463 DOI: 10.1186/s12885-018-4329-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 04/03/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Knowledge on periprosthetic infection and mortality rate following total knee arthroplasty (TKA) is essential for justifying this treatment in patients with cancer; however, relevant data from population-based studies are lacking. Therefore, we examined 1-year periprosthetic infection, mortality, and 5-year relative survival rates in cancer patients who underwent TKA. METHODS This is a population-based cohort study based on analysis of the Taiwan National Health Insurance Research Database. We enrolled a total of 2294 cancer patients and 131,849 patients without cancer (control group) who underwent TKA between January 1, 1997, and December 31, 2011. All patients were followed until death, infection, withdrawal from the National Health Insurance, or December 31, 2012. RESULTS The periprosthetic knee joint infection rate in cancer patients (1.73%) was not significantly higher than that in the control group (1.87%). However, the 1-year mortality rate was significantly higher (p < 0.05) in the cancer group (4.10%) than in the control group (1.66%). The overall 5-year survival rate was 93.10% as compared with those without cancers. CONCLUSION Low periprosthetic knee joint infection rates and high 5-year relative survival rates indicate the feasibility of TKA in cancer patients. However, the surgeon should take into account a higher mortality rate in the first year following TKA.
Collapse
Affiliation(s)
- Feng-Chen Kao
- Department of Orthopaedics, E-Da Hospital, Kaohsiung, Taiwan.,School of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Yao-Chun Hsu
- School of Medicine, Fu-Jen Catholic University, New Taipei, Taiwan. .,Department of Internal Medicine, E-Da Hospital, Kaohsiung, Taiwan. .,Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan. .,Division of Gastroenterology and Hepatology, Fu-Jen Catholic University Hospital, New Taipei, Taiwan.
| | - Pang-Yu Lai
- Department of Oncology, E-Da Hospital, Kaohsiung, Taiwan
| | - Chang-Bi Wang
- Graduate Institute of Public Health, China Medical University, Taichung, Taiwan.,Department of Statistics, National Taipei University, Taipei, Taiwan
| | - Yuan-Kun Tu
- Department of Orthopaedics, E-Da Hospital, Kaohsiung, Taiwan.,School of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Wen-Kang Chen
- Department of Applied Cosmetology, National Tainan Junior College of Nursing, Tainan, Taiwan
| |
Collapse
|
42
|
Blumenthal DT, Gorlia T, Gilbert MR, Kim MM, Burt Nabors L, Mason WP, Hegi ME, Zhang P, Golfinopoulos V, Perry JR, Hyun Nam D, Erridge SC, Corn BW, Mirimanoff RO, Brown PD, Baumert BG, Mehta MP, van den Bent MJ, Reardon DA, Weller M, Stupp R. Is more better? The impact of extended adjuvant temozolomide in newly diagnosed glioblastoma: a secondary analysis of EORTC and NRG Oncology/RTOG. Neuro Oncol 2018; 19:1119-1126. [PMID: 28371907 DOI: 10.1093/neuonc/nox025] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Background Radiation with concurrent and adjuvant (6 cycles) temozolomide (TMZ) is the established standard of postsurgical care for newly diagnosed glioblastoma (GBM). This regimen has been adopted with variations, including extending TMZ beyond 6 cycles. The optimal duration of maintenance therapy remains controversial. Methods We performed pooled analysis of individual patient data from 4 randomized trials for newly diagnosed GBM. All patients who were progression free 28 days after cycle 6 were included. The decision to continue TMZ was per local practice and standards, and at the discretion of the treating physician. Patients were grouped into those treated with 6 cycles and those who continued beyond 6 cycles. Progression-free and overall survival were compared, adjusted by age, performance status, resection extent, and MGMT methylation. Results A total of 2214 GBM patients were included in the 4 trials. Of these, 624 qualified for analysis 291 continued maintenance TMZ until progression or up to 12 cycles, while 333 discontinued TMZ after 6 cycles. Adjusted for prognostic factors, treatment with more than 6 cycles of TMZ was associated with a somewhat improved progression-free survival (hazard ratio [HR] 0.80 [0.65-0.98], P = .03), in particular for patients with methylated MGMT (n = 342, HR 0.65 [0.50-0.85], P < .01). However, overall survival was not affected by the number of TMZ cycles (HR = 0.92 [0.71-1.19], P = .52), including the MGMT methylated subgroup (HR = 0.89 [0.63-1.26], P = .51). Conclusions Continuing TMZ beyond 6 cycles was not shown to increase overall survival for newly diagnosed GBM.
Collapse
Affiliation(s)
- Deborah T Blumenthal
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Thierry Gorlia
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Mark R Gilbert
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Michelle M Kim
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - L Burt Nabors
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Warren P Mason
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Monika E Hegi
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Peixin Zhang
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Vassilis Golfinopoulos
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - James R Perry
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Do Hyun Nam
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Sara C Erridge
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Benjamin W Corn
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - René O Mirimanoff
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Paul D Brown
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Brigitta G Baumert
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Minesh P Mehta
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Martin J van den Bent
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - David A Reardon
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Michael Weller
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| | - Roger Stupp
- Tel-Aviv Sourasky Medical Center, Tel-Aviv University; European Organization for Research and Treatment of Cancer, Brussels (EORTC); National Institutes of Health (M.R.M.); University of Alabama at Birmingham; Princess Margaret Cancer Centre, University of Toronto; Lausanne University Hospital; NRG Oncology Statistics and Data Management Center; Odette Cancer Centre and Sunnybrook Health Sciences Centre, University of Toronto; Samsung Medical Center, Sungkyunkwan University School of Medicine; Edinburgh Cancer Centre; Mayo Clinic; Robert-Janker Clinic at the University of Bonn Medical Centre, and MAASTRO clinic, GROW School for Oncology, Maastricht University Medical Centre; Miami Cancer Institute; Erasmus University Hospital; Dana-Farber Cancer Institute and Harvard Medical School; University of Zurich
| |
Collapse
|
43
|
Tena I, Gupta G, Tajahuerce M, Benavent M, Cifrián M, Falcon A, Fonfria M, Del Olmo M, Reboll R, Conde A, Moreno F, Balaguer J, Cañete A, Palasí R, Bello P, Marco A, Ponce JL, Merino JF, Llombart A, Sanchez A, Pacak K. Successful Second-Line Metronomic Temozolomide in Metastatic Paraganglioma: Case Reports and Review of the Literature. CLINICAL MEDICINE INSIGHTS-ONCOLOGY 2018; 12:1179554918763367. [PMID: 29720885 PMCID: PMC5922490 DOI: 10.1177/1179554918763367] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 02/04/2018] [Indexed: 12/25/2022]
Abstract
Metastatic pheochromocytoma and paraganglioma (mPHEO/PGL) are frequently associated with succinate dehydrogenase B (SDHB) mutations. Cyclophosphamide-dacarbazine-vincristine (CVD) regimen is recommended as standard chemotherapy for advanced mPHEO/PGL. There is limited evidence to support the role of metronomic schemes (MS) of chemotherapy in mPHEO/PGL treatment. We report 2 patients with SDHB-related mPGL who received a regimen consisting of MS temozolomide (TMZ) and high-dose lanreotide after progression on both CVD chemotherapy and high-dose lanreotide. Molecular profiling of the tumor tissue from both patients revealed hypermethylation of the O6-methylguanine-DNA-methyltransferase (MGMT) promoter. In one patient, progression-free survival was 13 months and the second patient remained under treatment after 27 months of stabilization of metabolic response of his disease. Treatment was well tolerated, and adverse effects were virtually absent. A modification in the scheme of TMZ from standard schemes to MS is safe and feasible and can be considered in patients with progressive mPHEO/PGL refractory to dacarbazine in standard doses.
Collapse
Affiliation(s)
- Isabel Tena
- Department of Medical Oncology, Castellon Provincial Hospital, Castellón, Spain.,Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Garima Gupta
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Marcos Tajahuerce
- Department of Medical Oncology, Castellon Provincial Hospital, Castellón, Spain
| | - Marta Benavent
- Medical Oncology Department, Virgen del Rocío University Hospital, Seville, Spain
| | | | - Alejandro Falcon
- Medical Oncology Department, Virgen del Rocío University Hospital, Seville, Spain
| | - María Fonfria
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | | | - Rosa Reboll
- Department of Medical Oncology, Arnau de Vilanova Hospital, Valencia, Spain
| | - Antonio Conde
- Department of Medical Oncology, Castellon Provincial Hospital, Castellón, Spain
| | | | | | | | | | | | | | | | | | - Antonio Llombart
- Department of Medical Oncology, Arnau de Vilanova Hospital, Valencia, Spain
| | - Alfredo Sanchez
- Department of Medical Oncology, Castellon Provincial Hospital, Castellón, Spain
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
44
|
Zhang C, Li J, Wang H, Song SW. Identification of a five B cell-associated gene prognostic and predictive signature for advanced glioma patients harboring immunosuppressive subtype preference. Oncotarget 2018; 7:73971-73983. [PMID: 27738332 PMCID: PMC5342028 DOI: 10.18632/oncotarget.12605] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 09/27/2016] [Indexed: 12/29/2022] Open
Abstract
High grade gliomas contribute to most brain tumor mortality. A few studies reported that the immune system affected glioma development, and immune biomarkers helped understand the disease and formulate effective immunotherapy for patients. Currently, no B lymphocyte-based prognostic signature was reported in gliomas. By applying 78 B cell lineage-specific genes, we conducted a whole-genome gene expression analysis in 782 high grade gliomas derived from three independent datasets by Cox regression analysis and risk score method for signature identification, and then used Gene Ontology, Gene Set Enrichment Analysis, and other statistical methods for functional annotations of the signature-defined differences. We developed a five B cell-associated gene signature for prognosis of high grade glioma patients, which is independent of clinicopathological and genetic features. The signature identified high risk patients suitable for chemoradiotherapy, whereas low risk patients should rule out chemotherapy with radiotherapy only. We found that tumors of TCGA Mesenchymal subtype and wild type IDH1 were preferentially stratified to the high risk group, which bore strong immunosuppressive microenvironment, while tumors of TCGA Proneural subtype and mutated IDH1 were significantly accumulated to the low risk group, which exhibited less immunosuppressive state. The five B cell-associated gene signature predicts poor survival of high risk patients bearing strong immunosuppression and helps select optimal therapeutic regimens for glioma patients.
Collapse
Affiliation(s)
- Chuanbao Zhang
- Beijing Neurosurgical Institute, Capital Medical University, TiantanXili, Dongcheng District, Beijing 100050, China
| | - Jiye Li
- Beijing Neurosurgical Institute, Capital Medical University, TiantanXili, Dongcheng District, Beijing 100050, China.,Beijing Institute for Brain Disorders, Youanmen, Beijing, 100069, China.,Center for Brain Disorders Research, Capital Medical University, Youanmen, Beijing, 100069, China
| | - Haoyuan Wang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Sonya Wei Song
- Beijing Neurosurgical Institute, Capital Medical University, TiantanXili, Dongcheng District, Beijing 100050, China.,Beijing Institute for Brain Disorders, Youanmen, Beijing, 100069, China.,Center for Brain Disorders Research, Capital Medical University, Youanmen, Beijing, 100069, China
| |
Collapse
|
45
|
Suryadevara CM, Desai R, Abel ML, Riccione KA, Batich KA, Shen SH, Chongsathidkiet P, Gedeon PC, Elsamadicy AA, Snyder DJ, Herndon JE, Healy P, Archer GE, Choi BD, Fecci PE, Sampson JH, Sanchez-Perez L. Temozolomide lymphodepletion enhances CAR abundance and correlates with antitumor efficacy against established glioblastoma. Oncoimmunology 2018; 7:e1434464. [PMID: 29872570 PMCID: PMC5980382 DOI: 10.1080/2162402x.2018.1434464] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 01/24/2018] [Accepted: 01/25/2018] [Indexed: 11/24/2022] Open
Abstract
Adoptive transfer of T cells expressing chimeric antigen receptors (CARs) is an effective immunotherapy for B-cell malignancies but has failed in some solid tumors clinically. Intracerebral tumors may pose challenges that are even more significant. In order to devise a treatment strategy for patients with glioblastoma (GBM), we evaluated CARs as a monotherapy in a murine model of GBM. CARs exhibited poor expansion and survival in circulation and failed to treat syngeneic and orthotopic gliomas. We hypothesized that CAR engraftment would benefit from host lymphodepletion prior to immunotherapy and that this might be achievable by using temozolomide (TMZ), which is standard treatment for these patients and has lymphopenia as its major side effect. We modelled standard of care temozolomide (TMZSD) and dose-intensified TMZ (TMZDI) in our murine model. Both regimens are clinically approved and provide similar efficacy. Only TMZDI pretreatment prompted dramatic CAR proliferation and enhanced persistence in circulation compared to treatment with CARs alone or TMZSD + CARs. Bioluminescent imaging revealed that TMZDI + CARs induced complete regression of 21-day established brain tumors, which correlated with CAR abundance in circulation. Accordingly, TMZDI + CARs significantly prolonged survival and led to long-term survivors. These findings are highly consequential, as it suggests that GBM patients may require TMZDI as first line chemotherapy prior to systemic CAR infusion to promote CAR engraftment and antitumor efficacy. On this basis, we have initiated a phase I trial in patients with newly diagnosed GBM incorporating TMZDI as a preconditioning regimen prior to CAR immunotherapy (NCT02664363).
Collapse
Affiliation(s)
- Carter M. Suryadevara
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- Department of Pathology, Duke University Medical Center, Durham, NC
| | - Rupen Desai
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC
| | - Melissa L. Abel
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC
| | - Katherine A. Riccione
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- Department of Biomedical Engineering, Duke University, Durham, NC
| | - Kristen A. Batich
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- Department of Pathology, Duke University Medical Center, Durham, NC
| | - Steven H. Shen
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- Department of Pathology, Duke University Medical Center, Durham, NC
| | - Pakawat Chongsathidkiet
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- Department of Pathology, Duke University Medical Center, Durham, NC
| | - Patrick C. Gedeon
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- Department of Pathology, Duke University Medical Center, Durham, NC
| | - Aladine A. Elsamadicy
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC
| | - David J. Snyder
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC
| | - James E. Herndon
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC
- Duke Cancer Institute Biostatistics, Duke University Medical Center, Durham, NC
| | - Patrick Healy
- Duke Cancer Institute Biostatistics, Duke University Medical Center, Durham, NC
| | - Gary E. Archer
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- Department of Pathology, Duke University Medical Center, Durham, NC
| | - Bryan D. Choi
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- Department of Pathology, Duke University Medical Center, Durham, NC
| | - Peter E. Fecci
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- Department of Pathology, Duke University Medical Center, Durham, NC
| | - John H. Sampson
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- Department of Pathology, Duke University Medical Center, Durham, NC
- Department of Biomedical Engineering, Duke University, Durham, NC
| | - Luis Sanchez-Perez
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC
| |
Collapse
|
46
|
Kamran N, Alghamri MS, Nunez FJ, Shah D, Asad AS, Candolfi M, Altshuler D, Lowenstein PR, Castro MG. Current state and future prospects of immunotherapy for glioma. Immunotherapy 2018; 10:317-339. [PMID: 29421984 PMCID: PMC5810852 DOI: 10.2217/imt-2017-0122] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 11/30/2017] [Indexed: 12/14/2022] Open
Abstract
There is a large unmet need for effective therapeutic approaches for glioma, the most malignant brain tumor. Clinical and preclinical studies have enormously expanded our knowledge about the molecular aspects of this deadly disease and its interaction with the host immune system. In this review we highlight the wide array of immunotherapeutic interventions that are currently being tested in glioma patients. Given the molecular heterogeneity, tumor immunoediting and the profound immunosuppression that characterize glioma, it has become clear that combinatorial approaches targeting multiple pathways tailored to the genetic signature of the tumor will be required in order to achieve optimal therapeutic efficacy.
Collapse
Affiliation(s)
- Neha Kamran
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
- Department of Cell & Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
| | - Mahmoud S Alghamri
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
- Department of Cell & Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
| | - Felipe J Nunez
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
- Department of Cell & Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
| | - Diana Shah
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
- Department of Cell & Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
| | - Antonela S Asad
- Instituto de Investigaciones Biomédicas (CONICET-UBA), Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Marianela Candolfi
- Instituto de Investigaciones Biomédicas (CONICET-UBA), Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - David Altshuler
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
- Department of Cell & Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
| | - Pedro R Lowenstein
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
- Department of Cell & Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
| | - Maria G Castro
- Department of Neurosurgery, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
- Department of Cell & Developmental Biology, The University of Michigan School of Medicine, MSRB II, RM 4570C, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689, USA
| |
Collapse
|
47
|
Liposomal TriCurin, A Synergistic Combination of Curcumin, Epicatechin Gallate and Resveratrol, Repolarizes Tumor-Associated Microglia/Macrophages, and Eliminates Glioblastoma (GBM) and GBM Stem Cells. Molecules 2018; 23:molecules23010201. [PMID: 29346317 PMCID: PMC6017476 DOI: 10.3390/molecules23010201] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/03/2018] [Accepted: 01/15/2018] [Indexed: 01/10/2023] Open
Abstract
Glioblastoma (GBM) is a deadly brain tumor with a current mean survival of 12-15 months. Despite being a potent anti-cancer agent, the turmeric ingredient curcumin (C) has limited anti-tumor efficacy in vivo due to its low bioavailability. We have reported earlier a strategy involving the use two other polyphenols, epicatechin gallate (E) from green tea and resveratrol (R) from red grapes at a unique, synergistic molar ratio with C (C:E:R: 4:1:12.5, termed TriCurin) to achieve superior potency against HPV+ tumors than C alone at C:E:R (μM): 32:8:100 (termed 32 μM+ TriCurin). We have now prepared liposomal TriCurin (TrLp) and demonstrated that TrLp boosts activated p53 in cultured GL261 mouse GBM cells to trigger apoptosis of GBM and GBM stem cells in vitro. TrLp administration into mice yielded a stable plasma concentration of 210 nM C for 60 min, which, though sub-lethal for cultured GL261 cells, was able to cause repolarization of M2-like tumor (GBM)-associated microglia/macrophages to the tumoricidal M1-like phenotype and intra-GBM recruitment of activated natural killer cells. The intratumor presence of such tumoricidal immune cells was associated with concomitant suppression of tumor-load, and apoptosis of GBM and GBM stem cells. Thus, TrLp is a potential onco-immunotherapeutic agent against GBM tumors.
Collapse
|
48
|
Wenger A, Werlenius K, Hallner A, Thorén FB, Farahmand D, Tisell M, Smits A, Rydenhag B, Jakola AS, Carén H. Determinants for Effective ALECSAT Immunotherapy Treatment on Autologous Patient-Derived Glioblastoma Stem Cells. Neoplasia 2017; 20:25-31. [PMID: 29190492 PMCID: PMC5715204 DOI: 10.1016/j.neo.2017.10.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 10/24/2017] [Accepted: 10/24/2017] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive primary brain tumor with a median survival of less than 15 months, emphasizing the need for better treatments. Immunotherapy as a treatment for improving or aiding the patient's own immune defense to target the tumor has been suggested for GBM. A randomized clinical trial of adoptive cell transfer using ALECSAT (Autologous Lymphoid Effector Cells Specific Against Tumor Cells) is currently ongoing in Sweden. Here we performed a paired pre-clinical study to investigate the composition and in vitro effect of ALECSAT and identify determinants for the effect using autologous GBM-derived cancer stem cells (CSC), immunocytochemistry and flow cytometry. We show a clear dose-response relationship of ALECSAT on CSC, suggesting that the number of infused cells is of importance. In addition, the in vitro effect of ALECSAT on CSC correlated significantly to the blood count of T helper (Th) cells in the patient indicating a potential benefit of collecting cells for ALECSAT preparation at an even earlier stage when patients generally have a better blood count. The factors identified in this study will be important to consider in the design of future immunotherapy trials to achieve prolonged survival.
Collapse
Affiliation(s)
- Anna Wenger
- Sahlgrenska Cancer Center, Department of Pathology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Katja Werlenius
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden; Sahlgrenska Cancer Center, Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Alexander Hallner
- TIMM laboratory, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - Fredrik Bergh Thorén
- TIMM laboratory, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - Dan Farahmand
- Department of Neurosurgery, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Magnus Tisell
- Department of Neurosurgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anja Smits
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Neuroscience, Neurology, Uppsala University, University Hospital, Uppsala, Sweden
| | - Bertil Rydenhag
- Department of Neurosurgery, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Asgeir S Jakola
- Department of Neurosurgery, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Neurosurgery, St. Olavs University Hospital, Trondheim, Norway
| | - Helena Carén
- Sahlgrenska Cancer Center, Department of Pathology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden.
| |
Collapse
|
49
|
Lin AJ, Campian JL, Hui C, Rudra S, Rao YJ, Thotala D, Hallahan D, Huang J. Impact of concurrent versus adjuvant chemotherapy on the severity and duration of lymphopenia in glioma patients treated with radiation therapy. J Neurooncol 2017; 136:403-411. [PMID: 29143923 DOI: 10.1007/s11060-017-2668-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 11/11/2017] [Indexed: 11/24/2022]
Abstract
Prolonged severe lymphopenia has been shown to persist beyond a year in glioma patients after radiation therapy (RT) with concurrent and adjuvant chemotherapy. This study examines the differential impact of concurrent versus adjuvant chemotherapy on lymphopenia after RT. WHO grade II-III glioma patients who received RT with concurrent and/or adjuvant chemotherapy from 2007 to 2016 were retrospectively analyzed. Concurrent chemotherapy was temozolomide (TMZ), and adjuvant chemotherapy was either TMZ or procarbazine/lomustine/vincristine (PCV). Absolute lymphocyte count (ALC) was analyzed at baseline, 1.5, 3, 6, and 12 months after the start of RT. Univariable and multivariable logistic regression were used to identify the clinical variables in predicting acute or late lymphopenia. There were 151 patients with evaluable ALC: 91 received concurrent and adjuvant TMZ (CRT + ADJ), 32 received only concurrent TMZ (CRT), and 28 received only adjuvant TMZ or PCV (ADJ). There were 9 (10%) versus 6 (19%) versus 0 (0%) cases of grade 3 lymphopenia (ALC < 500/mm3) at 6 weeks and 4 (6%) versus 0 (0%) versus 3 (17%) cases at 12 months in CRT + ADJ, CRT and ADJ groups, respectively. On multivariable analyses, concurrent chemotherapy (odds ratio [OR] 72.3, p < 0.001), female sex (OR 10.8, p < 0.001), and older age (OR 1.06, p = 0.002) were the most significant predictors for any grade ≥ 1 lymphopenia (ALC < 1000/mm3) at 1.5 months. Older age (OR 1.08, p = 0.02) and duration of adjuvant chemotherapy (OR 1.19, p = 0.003) were significantly associated with grade ≥ 1 lymphopenia at 12 months. Thus, concurrent chemotherapy appears as the dominant contributor to the severity of acute lymphopenia after RT in WHO grade II-III glioma patients, and duration of adjuvant chemotherapy appears as the key factor to prolonged lymphopenia.
Collapse
Affiliation(s)
- Alexander J Lin
- Department of Radiation Oncology, Center for Advanced Medicine, Washington University School of Medicine, 4921 Parkview Place, Campus Box #8224, St. Louis, MO, 63110, USA
| | - Jian L Campian
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Caressa Hui
- Department of Radiation Oncology, Center for Advanced Medicine, Washington University School of Medicine, 4921 Parkview Place, Campus Box #8224, St. Louis, MO, 63110, USA
| | - Soumon Rudra
- Department of Radiation Oncology, Center for Advanced Medicine, Washington University School of Medicine, 4921 Parkview Place, Campus Box #8224, St. Louis, MO, 63110, USA
| | - Yuan J Rao
- Department of Radiation Oncology, Center for Advanced Medicine, Washington University School of Medicine, 4921 Parkview Place, Campus Box #8224, St. Louis, MO, 63110, USA
| | - Dinesh Thotala
- Department of Radiation Oncology, Center for Advanced Medicine, Washington University School of Medicine, 4921 Parkview Place, Campus Box #8224, St. Louis, MO, 63110, USA
| | - Dennis Hallahan
- Department of Radiation Oncology, Center for Advanced Medicine, Washington University School of Medicine, 4921 Parkview Place, Campus Box #8224, St. Louis, MO, 63110, USA
| | - Jiayi Huang
- Department of Radiation Oncology, Center for Advanced Medicine, Washington University School of Medicine, 4921 Parkview Place, Campus Box #8224, St. Louis, MO, 63110, USA.
| |
Collapse
|
50
|
Luksik AS, Maxwell R, Garzon-Muvdi T, Lim M. The Role of Immune Checkpoint Inhibition in the Treatment of Brain Tumors. Neurotherapeutics 2017; 14:1049-1065. [PMID: 28258545 PMCID: PMC5722751 DOI: 10.1007/s13311-017-0513-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The standard of care for malignant gliomas of the brain has changed very little over the last few decades, and does not offer a cure for these rare, but fatal, tumors. The field of immunotherapy has brought potent new drugs into the oncological armamentarium, and is becoming recognized as a potentially important arm in the treatment of glioblastoma for adults. Immune checkpoints are inhibitory receptors found on immune cells that, when stimulated, cause those immune cells to become quiescent. While this is a natural mechanism to prevent excessive inflammatory damage and autoimmunity in otherwise healthy tissues, cancer cells may utilize this process to grow in the absence of targeted immune destruction. Antibodies derived to block the stimulation of these negative checkpoints, allowing immune cells to remain activated and undergo effector function, are a growing area of immunotherapy. These therapies have seen much success in both the preclinical and clinical arenas for various tumors, particularly melanoma and nonsmall-cell lung cancer. Multiple clinical trials are underway to determine if these drugs have efficacy in glioblastoma. Here, we review the current evidence, from early preclinical data to lessons learned from clinical trials outside of glioblastoma, to assess the potential of immune checkpoint inhibition in the treatment of brain tumors and discuss how this therapy may be implemented with the present standard of care.
Collapse
Affiliation(s)
- Andrew S Luksik
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Russell Maxwell
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tomas Garzon-Muvdi
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael Lim
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|