1
|
Buckley MW, Balaji Warner A, Brahmer J, Cappelli LC, Sharfman WH, Fuchs E, Kang H, Forde PM, Gladstone DE, Ambinder R, Kelly RJ, Lipson EJ, Gojo I, Lee EJ, Johnson TP, Saidha S, Llinas R, Ostrow LW, Naidoo J, Probasco JC. Immune-related encephalitis after immune checkpoint inhibitor therapy. Oncologist 2025; 30:oyae186. [PMID: 39066587 PMCID: PMC11783331 DOI: 10.1093/oncolo/oyae186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI) have revolutionized cancer treatment but can trigger immune-related encephalitis. We report one of the largest case series of patients with immune-related encephalitis and review of the literature. METHODS Retrospective series of patients with immune-related encephalitis and literature review. RESULTS Fourteen patients with cancer treated with ICI (50% combination therapy) developed immune-related encephalitis. Diagnostic testing revealed cerebral spinal fluid (CSF) lymphocytic pleocytosis (85%) and elevated protein (69%), abnormal brain magnetic resonance imaging(MRI) (33%) or brain FDG-PET (25%), electroencephalogram (EEG) abnormalities (30%), and autoantibodies (31%). Encephalitis treatment included: corticosteroids (86%), intravenous immunoglobulin (IVIg) (36%), plasmapheresis (7%), and rituximab (29%). There were no deaths and 12 patients had significant recovery, although long-term complications were observed. All patients discontinued ICI. Longitudinal follow-up demonstrated anti-cancer response to ICI at 3 months (85%) and 6 months post-ICI initiation (77%). A literature review identified 132 patients with immune-related encephalitis. Most were treated with PD-1 inhibitors (18% combination). Common abnormalities included elevated CSF protein (84%) or pleocytosis (77%), abnormal brain MRI (65%), or autoantibodies (47%). Nearly all were treated with corticosteroids, many required additional therapy with IVIg (26%) or rituximab (12%). Most patients had clinical improvement (81%) but a minority (10%) had a clinical relapse after completing corticosteroid taper. ICIs were resumed in 7 patients (5%), with relapse in 3. CONCLUSIONS AND RELEVANCE Immune-related encephalitis is treatable and improves with corticosteroids in most cases but may require additional immunosuppression. Re-emergence of encephalitis is rare and does not typically result in adverse outcomes, and this should be considered in neurological immune-related adverse event management guidelines.
Collapse
Affiliation(s)
- Monica W Buckley
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Department of Neurology, University of Virginia School of Medicine, Charlottesville, VA 22903, United States
| | - Aanika Balaji Warner
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287, United States
| | - Julie Brahmer
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287, United States
| | - Laura C Cappelli
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - William H Sharfman
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287, United States
| | - Ephraim Fuchs
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Hyunseok Kang
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, United States
| | - Patrick M Forde
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287, United States
| | - Douglas E Gladstone
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- R.J. Zuckerberg Cancer Center at Hofstra/Northwell Health, Lake Success, NY 11042, United States
| | - Richard Ambinder
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Ronan J Kelly
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, TX 75246, United States
| | - Evan J Lipson
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287, United States
| | - Ivana Gojo
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Edward J Lee
- Maryland Oncology Hematology, Columbia, MD 21044, United States
| | - Tory P Johnson
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Shiv Saidha
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Rafael Llinas
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Lyle W Ostrow
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Department of Neurology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Jarushka Naidoo
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287, United States
- Department of Oncology, Johns Hopkins Bayview Medical Center, Baltimore, MD 21224, United States
- Department of Medicine, Beaumont Hospital Dublin and RCSI University of Health Sciences, Dublin, 9, Ireland
| | - John C Probasco
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| |
Collapse
|
2
|
Grabbe P, Borchers MS, Gschwendtner KM, Strobel S, Wild B, Kirchner M, Kälber K, Rendon A, Steininger J, Meier F, Hassel JC, Bieber C. An Online Decision Aid for Patients With Metastatic Melanoma—Results of the Randomized Controlled Trial “PEF-Immun”. DEUTSCHES ARZTEBLATT INTERNATIONAL 2024; 121:385-392. [PMID: 38566437 PMCID: PMC11460262 DOI: 10.3238/arztebl.m2024.0053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 03/11/2024] [Accepted: 03/11/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND Treatment decisions in metastatic melanoma (MM) are highly dependent on patient preferences and require the patients' involvement. The complexity of treatment options with their individual advantages and disadvantages is often overwhelming. We therefore developed an online patient decision aid (PtDA) to facilitate shared decision making (SDM). METHODS To evaluate the PtDA we conducted a two-armed, twocenter, prospective, open randomized controlled trial with MM patients who were facing a decision about first-line treatment. The patients were allotted randomly in a 1:1 ratio to an intervention group (IG) with access to the PtDA before discussion with a physician or to a control group (CG) without access to the PtDA. The primary endpoint was knowledge about the options for first-line treatment (multiple-choice test, 10 items, range 0-40 points). The secondary endpoints were the SDM (third-party ratings of audio recordings of the treatment discussions) and satisfaction with the decision at the follow-up visit. RESULTS Of the 128 randomized patients, 120 completed the baseline questionnaire and were analyzed (59% male, median age 66 years). The primary endpoint, i.e., the mean difference in knowledge after discussion with a physician, differed significantly between the IG and the CG (-3.22, 95% CI [-6.32; -0.12], p = 0.042). No differences were found for the secondary endpoints, SDM and satisfaction with the decision. The patients in the IG rated the PtDA as very useful. CONCLUSION The PtDA improved the knowledge of patients with MM about the options for treatment. Both groups were highly satisfied with their treatment decisions. However, additional physician training seems necessary to promote SDM.
Collapse
Affiliation(s)
- Pia Grabbe
- *Joint first authors
- Department of General Internal Medicine and Psychosomatics, Center for Psychosocial Medicine, Heidelberg University Hospital, Medical Faculty, Heidelberg University, Heidelberg
| | - Milena S. Borchers
- *Joint first authors
- Department of General Internal Medicine and Psychosomatics, Center for Psychosocial Medicine, Heidelberg University Hospital, Medical Faculty, Heidelberg University, Heidelberg
| | - Kathrin M. Gschwendtner
- Department of General Internal Medicine and Psychosomatics, Center for Psychosocial Medicine, Heidelberg University Hospital, Medical Faculty, Heidelberg University, Heidelberg
| | - Sophia Strobel
- Heidelberg University, Medical Faculty, Department of Dermatology and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
| | - Beate Wild
- Department of General Internal Medicine and Psychosomatics, Center for Psychosocial Medicine, Heidelberg University Hospital, Medical Faculty, Heidelberg University, Heidelberg
| | - Marietta Kirchner
- Institute of Medical Biometry, Heidelberg University Hospital, Heidelberg
| | - Katharina Kälber
- Heidelberg University, Medical Faculty, Department of Dermatology and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
| | - Adriana Rendon
- Heidelberg University, Medical Faculty, Department of Dermatology and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
| | - Julian Steininger
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus at the Technical University Dresden, Dresden, Germany
| | - Friedegund Meier
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus at the Technical University Dresden, Dresden, Germany
| | - Jessica C. Hassel
- *Joint last authors
- Heidelberg University, Medical Faculty, Department of Dermatology and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
| | | |
Collapse
|
3
|
Bi S, Chen W, Fang Y, Shen J, Zhang Q, Guo H, Ju H, Liu Y. Cancer Cell-Selective PD-L1 Inhibition via a DNA Safety Catch to Enhance Immunotherapy Specificity. Angew Chem Int Ed Engl 2024; 63:e202402522. [PMID: 38421189 DOI: 10.1002/anie.202402522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 02/29/2024] [Accepted: 02/29/2024] [Indexed: 03/02/2024]
Abstract
Immune checkpoint protein blockade (ICB) has emerged as a powerful immunotherapy approach, but suppressing immune-related adverse events (irAEs) for noncancerous cells and normal tissues remains challenging. Activatable ICB has been developed with tumor microenvironment highly-expressed molecules as stimuli, but they still lack precision and efficiency considering the diffusion of stimuli molecules in whole tumor tissue. Here we assemble PD-L1 with a duplex DNA strand, termed as "safety catch", to regulate its accessibility for ICB. The safety catch remains at "on" status for noncancerous cells to prevent ICB binding to PD-L1. Cancer cell membrane protein c-Met acts as a trigger protein to react with safety catch, which selectively exposes its hybridization region for ICB reagent. The ICB reagent is a retractable DNA nanostring with repeating hairpin-structural units, whose contraction drives PD-L1 clustering with endocytosis-guided degradation. The safety catch, even remained at "safety on" status, is removed from the cell membrane via a DNA strand displacement reaction to minimize its influence on noncancerous cells. This strategy demonstrates selective and potent immunotherapeutic capabilities only against cancer cells both in vitro and in vivo, and shows effective suppression of irAEs in normal tissues, therefore would become a promising approach for precise immunotherapy in mice.
Collapse
Affiliation(s)
- Shiyi Bi
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Wei Chen
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing, 210008, China
| | - Yanyun Fang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Jieyu Shen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Qing Zhang
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing, 210008, China
| | - Hongqian Guo
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing, 210008, China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Ying Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210023, China
| |
Collapse
|
4
|
Huang C, Ren S, Chen Y, Liu A, Wu Q, Jiang T, Lv P, Song D, Hu F, Lan J, Sun L, Zheng X, Luo X, Chu Q, Jia K, Li Y, Wang J, Zou C, Hu J, Wang G. PD-L1 methylation restricts PD-L1/PD-1 interactions to control cancer immune surveillance. SCIENCE ADVANCES 2023; 9:eade4186. [PMID: 37235656 DOI: 10.1126/sciadv.ade4186] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 04/21/2023] [Indexed: 05/28/2023]
Abstract
Immune checkpoint inhibitors targeting programmed cell death protein 1 (PD-1) or programmed cell death 1 ligand 1 (PD-L1) have enabled some patients with cancer to experience durable, complete treatment responses; however, reliable anti-PD-(L)1 treatment response biomarkers are lacking. Our research found that PD-L1 K162 was methylated by SETD7 and demethylated by LSD2. Furthermore, PD-L1 K162 methylation controlled the PD-1/PD-L1 interaction and obviously enhanced the suppression of T cell activity controlling cancer immune surveillance. We demonstrated that PD-L1 hypermethylation was the key mechanism for anti-PD-L1 therapy resistance, investigated that PD-L1 K162 methylation was a negative predictive marker for anti-PD-1 treatment in patients with non-small cell lung cancer, and showed that the PD-L1 K162 methylation:PD-L1 ratio was a more accurate biomarker for predicting anti-PD-(L)1 therapy sensitivity. These findings provide insights into the regulation of the PD-1/PD-L1 pathway, identify a modification of this critical immune checkpoint, and highlight a predictive biomarker of the response to PD-1/PD-L1 blockade therapy.
Collapse
Affiliation(s)
- Changsheng Huang
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shengxiang Ren
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Yaqi Chen
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Anyi Liu
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qi Wu
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tao Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Panjing Lv
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Da Song
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Fuqing Hu
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jingqing Lan
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Li Sun
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xue Zheng
- Wuhan Blood Center, Wuhan 430030, China
| | - Xuelai Luo
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qian Chu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Keyi Jia
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Yan Li
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jun Wang
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Caicun Zou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Junbo Hu
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Guihua Wang
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
5
|
Grabbe P, Gschwendtner KM, Maatouk I, Strobel SB, Salzmann M, Bossert J, Eich W, Wild B, Meier F, Hassel JC, Bieber C. Development and validation of a web-based patient decision aid for immunotherapy for patients with metastatic melanoma: study protocol for a multicenter randomized trial. Trials 2021; 22:294. [PMID: 33879219 PMCID: PMC8056554 DOI: 10.1186/s13063-021-05234-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 03/29/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Patients with metastatic melanoma and their physicians are confronted with a complex decision regarding first-line therapy. Risks and benefits vary considerably between various treatment options. With this in mind, we aim to develop and evaluate a patient decision aid (PtDA) to inform patients about the risks and benefits of treatment options, namely, immunotherapy as monotherapy, immunotherapy as combination therapy, and treatment with BRAF/MEK inhibitors. We aim to test whether the use of this PtDA before medical consultation will increase patients' knowledge of treatment options and thus promote shared decision-making (SDM) and patient decision satisfaction. METHODS In total, 128 patients with metastatic melanoma from two German cancer centers will be randomized to the intervention group (IG), receiving access to the PtDA before medical consultation, or the control group (CG), receiving treatment as usual (TAU), i.e., medical consultation alone. There will be three major assessment points (before intervention, T0; after intervention, T1; and 3 months after intervention, T2). The main outcome is the patient's knowledge of their treatment options, measured by a self-developed, piloted multiple-choice test at T1. Secondary outcome measures will include the extent of SDM during medical consultation, assessed by Observer OPTION 5, and patient decision satisfaction, assessed by the Satisfaction with Decision Scale (SwD), at T1 and T2. DISCUSSION This trial will assess the effectiveness of a developed PtDA to enhance patient knowledge of treatment options for metastatic melanoma, SDM, and patient decision satisfaction. If the efficacy can be proven, the PtDA will be implemented nationwide in Germany to close a relevant gap in the education and care of patients with metastatic melanoma. TRIAL REGISTRATION ClinicalTrials.gov NCT04240717 . Registered on 27 January 2020.
Collapse
Affiliation(s)
- Pia Grabbe
- Department of General Internal Medicine and Psychosomatics, Center for Psychosocial Medicine, Heidelberg University Hospital, Thibautstraße 4, 69115 Heidelberg, Germany
| | - Kathrin M. Gschwendtner
- Department of General Internal Medicine and Psychosomatics, Center for Psychosocial Medicine, Heidelberg University Hospital, Thibautstraße 4, 69115 Heidelberg, Germany
| | - Imad Maatouk
- Department of General Internal Medicine and Psychosomatics, Center for Psychosocial Medicine, Heidelberg University Hospital, Thibautstraße 4, 69115 Heidelberg, Germany
| | - Sophia B. Strobel
- National Center for Tumor Diseases, University Hospital Heidelberg, Im Neuenheimer Feld 460, 69120 Heidelberg, Germany
| | - Martin Salzmann
- National Center for Tumor Diseases, University Hospital Heidelberg, Im Neuenheimer Feld 460, 69120 Heidelberg, Germany
| | - Julia Bossert
- Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln, Germany
- TAKEPART Media + Science, Köln, Germany
| | - Wolfgang Eich
- Department of General Internal Medicine and Psychosomatics, Center for Psychosocial Medicine, Heidelberg University Hospital, Thibautstraße 4, 69115 Heidelberg, Germany
| | - Beate Wild
- Department of General Internal Medicine and Psychosomatics, Center for Psychosocial Medicine, Heidelberg University Hospital, Thibautstraße 4, 69115 Heidelberg, Germany
| | - Friedegund Meier
- National Center for Tumor Diseases, University Hospital Dresden, Fetscherstraße 74, 01307 Dresden, Germany
| | - Jessica C. Hassel
- National Center for Tumor Diseases, University Hospital Heidelberg, Im Neuenheimer Feld 460, 69120 Heidelberg, Germany
| | - Christiane Bieber
- Department of General Internal Medicine and Psychosomatics, Center for Psychosocial Medicine, Heidelberg University Hospital, Thibautstraße 4, 69115 Heidelberg, Germany
| |
Collapse
|
6
|
Meng L, He X, Zhang X, Zhang X, Wei Y, Wu B, Li W, Li J, Xiao Y. Predicting the clinical outcome of melanoma using an immune-related gene pairs signature. PLoS One 2020; 15:e0240331. [PMID: 33031392 PMCID: PMC7544036 DOI: 10.1371/journal.pone.0240331] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/23/2020] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Melanoma is rare but dangerous skin cancer, and it can spread rather quickly in the advanced stages of the tumor. Abundant evidence suggests the relationship between tumor development and progression and the immune system. A robust gene risk model could provide an accurate prediction of clinical outcomes. The present study aimed to explore a robust signature of immune-related gene pairs (IRGPs) for estimating overall survival (OS) in malignant melanoma. METHODS Clinical and genetic data of skin cutaneous melanoma (SKCM) patients from The Cancer Genome Atlas (TCGA) was performed as a training dataset to identify candidate IRGPs for the prognosis of melanoma. Two independent datasets from the Gene Expression Omnibus (GEO) database (GSE65904) and TCGA dataset (TCGA-UVM) were selected for external validation. Univariate and multivariate Cox regression analyses were then performed to explore the prognostic power of the IRGPs signature and other clinical factors. CIBERSORTx was applied to estimate the fractions of infiltrated immune cells in bulk tumor tissues. RESULTS A signature consisted of 33 IRGPs was established which was significantly associated with patients' survival in the TCGA-SKCM dataset (P = 2.0×10-16, Hazard Ratio (HR) = 4.220 (2.909 to 6.122)). We found the IRGPs signature exhibited an independent prognostic factor in all the three independent cohorts in both the univariate and multivariate Cox analysis (P<0.01). The prognostic efficacy of the signature remained unaffected regardless of whether BRAF or NRAS was mutated. As expected, the results were verified in the GSE65904 dataset and the TCGA-UVM dataset. We found an apparent shorter OS in patients of the high-risk group in the GSE65904 dataset (P = 2.1×10-3; HR = 1.988 (1.309 to 3.020)). The trend in the results of the survival analysis in TCGA-UVM was as we expected, but the result was not statistically significant (P = 0.117, HR = 4.263 (1.407 to 12.91)). CD8 T cells, activated dendritic cells (DCs), regulatory T cells (Tregs), and activated CD4 memory T cells presented a significantly lower fraction in the high-risk group in the TCGA-SKCM dataset(P <0.01). CONCLUSION The results of the present study support the IRGPs signature as a promising marker for prognosis prediction in melanoma.
Collapse
Affiliation(s)
- Liangliang Meng
- Medical School of Chinese PLA, Beijing, China
- Department of Radiology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
- Department of Radiology, Chinese PAP Beijing Corps Hospital, Beijing, China
| | - Xiaoxi He
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Xiao Zhang
- Department of Radiology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Xiaobo Zhang
- Department of Radiology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yingtian Wei
- Department of Radiology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Bin Wu
- Department of Radiology, Chinese PAP Beijing Corps Hospital, Beijing, China
| | - Wei Li
- Department of Radiology, Chinese PAP Beijing Corps Hospital, Beijing, China
| | - Jing Li
- Department of Radiology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yueyong Xiao
- Department of Radiology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
7
|
Wang J, Li Z, Wang Z, Yu Y, Li D, Li B, Ding J. Nanomaterials for Combinational Radio–Immuno Oncotherapy. ADVANCED FUNCTIONAL MATERIALS 2020; 30. [DOI: 10.1002/adfm.201910676] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 03/09/2020] [Indexed: 08/29/2023]
Abstract
AbstractRadiotherapy, a clinically used local treatment modality of cancers, is regarded as a promising candidate to promote current immunotherapy through initiating an in situ vaccination effect and reprogramming the immunosuppressive microenvironment. The combination of radiotherapy and immunotherapy, referred to as combinational radio–immuno oncotherapy (CRIOT), elicits a synergistic antitumor effect based on the immunomodulatory properties of radiation. Unfortunately, current CRIOT accompanies low response rate and severe toxicity in clinical trials, thus limiting its application. To this end, various nanomaterials are being developed to sensitize radiotherapy or deliver immune agents, or both, to improve the unsatisfactory outcomes of CRIOT. Herein, enhanced antitumor efficacy of CRIOT with nanomaterials through the possible mechanisms of rejuvenation and activation of T cells, increased presentation of tumor‐related antigens, and inhibition of suppressive macrophages is presented, and the prospect of CRIOT in clinical practice is proposed.
Collapse
Affiliation(s)
- Juan Wang
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry Chinese Academy of Sciences 5625 Renmin Street Changchun 130022 P. R. China
- Department of Radiation Oncology Cancer Hospital of Shandong First Medical University 440 Jiyan Road Jinan 250117 P. R. China
| | - Zhongmin Li
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry Chinese Academy of Sciences 5625 Renmin Street Changchun 130022 P. R. China
- Department of Gastrointestinal, Colorectal, and Anal Surgery China–Japan Union Hospital of Jilin University 126 Xiantai Street Changchun 130012 P. R. China
| | - Zhongtang Wang
- Department of Radiation Oncology Cancer Hospital of Shandong First Medical University 440 Jiyan Road Jinan 250117 P. R. China
| | - Yonghua Yu
- Department of Radiation Oncology Cancer Hospital of Shandong First Medical University 440 Jiyan Road Jinan 250117 P. R. China
| | - Di Li
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry Chinese Academy of Sciences 5625 Renmin Street Changchun 130022 P. R. China
| | - Baosheng Li
- Department of Radiation Oncology Cancer Hospital of Shandong First Medical University 440 Jiyan Road Jinan 250117 P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry Chinese Academy of Sciences 5625 Renmin Street Changchun 130022 P. R. China
| |
Collapse
|
8
|
Quon PL, Xiao Y, Sorensen S, Monfared AAT. Economic Evaluation of Nivolumab Plus Ipilimumab Combination as First-Line Treatment for Patients with Advanced Melanoma in Canada. PHARMACOECONOMICS - OPEN 2019; 3:321-331. [PMID: 30617952 PMCID: PMC6710483 DOI: 10.1007/s41669-018-0112-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
OBJECTIVE Our objective was to evaluate the cost effectiveness of the combination of nivolumab and ipilimumab, referred to as "Regimen", as a first-line treatment for patients with advanced melanoma from the perspective of Canada's public healthcare system. METHODS We developed a partitioned-survival model (progression-free survival, post-progression survival, and death) to determine the clinical and economic outcomes of immunotherapy for advanced melanoma over a 20-year time horizon. Regimen was compared with nivolumab, ipilimumab, and pembrolizumab. Two treatment durations for pembrolizumab were considered: (1) maximum of 24 months or until progression or (2) no maximum duration, until progression. The model used data from CheckMate-067 (28 months' follow-up) for treatments involving nivolumab and ipilimumab. The efficacy of pembrolizumab was estimated using indirect comparisons. A scenario looking at the cost of subsequent treatments following disease progression was examined. RESULTS Regimen had better outcomes and was cost effective compared with all other immunotherapies at a threshold of $CAN100,000 per quality-adjusted life-year (QALY) gained. Compared with nivolumab and ipilimumab, the incremental cost-effectiveness ratios (ICERs) were $CAN47,119 and 66,750 per QALY, respectively. Compared with pembrolizumab with a treatment duration cap, the ICER was $CAN85,436. When assuming no duration cap, Regimen dominated pembrolizumab. With the inclusion of subsequent treatment costs following progression, Regimen's ICER improved compared with all other comparators. CONCLUSIONS Despite the advent of effective new therapies for advanced melanoma, prognosis remains poor for some patients. Compared with other immunotherapies, Regimen offers marked benefit and may be a cost-effective treatment option.
Collapse
Affiliation(s)
- Peter L. Quon
- Modeling and Simulation, Evidera, 7101 Wisconsin Avenue, Suite 1400, Bethesda, MD 20814 USA
| | - Ying Xiao
- Modeling and Simulation, Evidera, 7101 Wisconsin Avenue, Suite 1400, Bethesda, MD 20814 USA
| | - Sonja Sorensen
- Modeling and Simulation, Evidera, 7101 Wisconsin Avenue, Suite 1400, Bethesda, MD 20814 USA
| | - Amir Abbas Tahami Monfared
- Bristol-Myers Squibb, Montreal, QC Canada
- Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC Canada
| |
Collapse
|
9
|
The Dosage of the Derivative of Clostridium Ghonii (DCG) Spores Dictates Whether an IFN γ/IL-9 or a Strong IFN γ Response Is Elicited in TC-1 Tumour Bearing Mice. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1395138. [PMID: 31183361 PMCID: PMC6512072 DOI: 10.1155/2019/1395138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 01/04/2019] [Accepted: 01/15/2019] [Indexed: 11/24/2022]
Abstract
Background Anaerobic Clostridial spores (CG) cause significant oncolysis in hypoxic tumour microenvironment and result in tumour regression in both animal models and clinical trials. The immune mediated response plays a critical role in the antitumour effect by the anaerobic spore treatment. Method Human papillomavirus 16 E6/E7 transformed TC-1 tumour bearing mice were intravenously administered with low (1 × 108 CFU/kg) or high dosage (3 × 108 CFU/kg) of Derivative Clostridial spore (DCG). Results Intravenous administration of the derivative of Clostridial ghonii (DCG) spores leads to both tumour and systemic inflammatory responses characterized by increased IFNγ/IL-9 secreting T cells in the spleen and the tumour. Low numbers of antigen specific T cells (<20/106 spleen cells) in the spleen of the tumour bearing mice are also detected after intravenous DCG delivery. Interestingly, our results showed that a mixed IL-9/IFNγ secreting T cell response was induced when the tumour bearing mice received a low dose of DCG spore (1 × 108 CFU/kg), while a strong IFNγ response was elicited with a high dosage of DCG spore (3 × 108 CFU/kg). Conclusion The dosage of DCG spore will determine the types of the DCG induced immune responses.
Collapse
|
10
|
Yang MW, Fu XL, Jiang YS, Chen XJ, Tao LY, Yang JY, Huo YM, Liu W, Zhang JF, Liu PF, Liu Q, Hua R, Zhang ZG, Sun YW, Liu DJ. Clinical significance of programmed death 1/programmed death ligand 1 pathway in gastric neuroendocrine carcinomas. World J Gastroenterol 2019; 25:1684-1696. [PMID: 31011254 PMCID: PMC6465942 DOI: 10.3748/wjg.v25.i14.1684] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/05/2019] [Accepted: 03/16/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Recently, more and more studies have demonstrated the pivotal role of programmed death 1/programmed death ligand 1 (PD-1/PD-L1) pathway in the immune evasion of tumors from the host immune system. However, the role of PD-1/PD-L1 pathway in gastric neuroendocrine carcinomas (G-NECs) remains unknown.
AIM To investigate the expression of PD-1/PD-L1 and role of PD-1/PD-L1 pathway in G-NECs, which occur rarely but are highly malignant and clinically defiant.
METHODS We investigated the expression of PD-L1 on tumor cells and PD-1+, CD8+, and FOXP3+ T cell infiltration by immunohistochemistry in 43 resected G-NEC tissue specimens. The copy number alterations of PD-L1 were assessed by qRT-PCR.
RESULTS Most of the G-NECs tumor cells exhibited a near-uniform expression pattern of PD-L1, while some showed a tumor-stromal interface enhanced pattern. Of the 43 G-NECs, 21 (48.8%) were classified as a high PD-L1 expression group, and the high expression of PD-L1 was associated with poor overall survival (OS). The high expression of PD-L1 was correlated with abundant PD-1+ tumor infiltrating lymphocytes (TILs) instead of CD8+ TILs and FOXP3+ regulatory T cells (Tregs). Our analysis also suggested that the infiltration of CD8+ TILs tended to be a favorable factor for OS, although the difference did not reach the statistical significance (P = 0.065). Meanwhile, PD-L1 was significantly overexpressed in cases with copy number gain as compared with those without.
CONCLUSION Our data demonstrated for the first time that high expression of PD-L1 in G-NECs is associated with a poor prognosis, while the high expression may be due to the copy number variation of PD-L1 gene or stimulation of TILs. These results provide a basis for the immunotherapy targeting PD-1/PD-L1 pathway in G-NECs.
Collapse
Affiliation(s)
- Min-Wei Yang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xue-Liang Fu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yong-Sheng Jiang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xiao-Jing Chen
- Central Laboratory, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Ling-Ye Tao
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jian-Yu Yang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yan-Miao Huo
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Wei Liu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jun-Feng Zhang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Pei-Feng Liu
- Central Laboratory, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Qiang Liu
- Department of Pathology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Rong Hua
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Zhi-Gang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yong-Wei Sun
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - De-Jun Liu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
11
|
Narang P, Chen M, Sharma AA, Anderson KS, Wilson MA. The neoepitope landscape of breast cancer: implications for immunotherapy. BMC Cancer 2019; 19:200. [PMID: 30832597 PMCID: PMC6399957 DOI: 10.1186/s12885-019-5402-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 02/22/2019] [Indexed: 02/07/2023] Open
Abstract
Background Cancer immunotherapy with immune checkpoint blockade (CKB) is now standard of care for multiple cancers. The clinical response to CKB is associated with T cell immunity targeting cancer-induced mutations that generate novel HLA-binding epitopes (neoepitopes). Methods Here, we developed a rapid bioinformatics pipeline and filtering strategy, EpitopeHunter, to identify and prioritize clinically relevant neoepitopes from the landscape of somatic mutations. We used the pipeline to determine the frequency of neoepitopes from the TCGA dataset of invasive breast cancers. We predicted HLA class I-binding neoepitopes for 870 breast cancer samples and filtered the neoepitopes based on tumor transcript abundance. Results We found that the total mutational burden (TMB) was highest for triple-negative breast cancer, TNBC, (median = 63 mutations, range: 2–765); followed by HER-2(+) (median = 39 mutations, range: 1–1206); and lowest for ER/PR(+)HER-2(−) (median = 32 mutations, range: 1–2860). 40% of the nonsynonymous mutations led to the generation of predicted neoepitopes. The neoepitope load (NEL) is highly correlated with the mutational burden (R2 = 0.86). Conclusions Only half (51%) of the predicted neoepitopes are expressed at the RNA level (FPKM≥2), indicating the importance of assessing whether neoepitopes are transcribed. However, of all patients, 93% have at least one expressed predicted neoepitope, indicating that most breast cancer patients have the potential for neo-epitope targeted immunotherapy. Electronic supplementary material The online version of this article (10.1186/s12885-019-5402-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Pooja Narang
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ, 85287-4501, USA
| | - Meixuan Chen
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ, 85287-4501, USA.,Center for Personalized Diagnostics, The Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Amit A Sharma
- Center for Personalized Diagnostics, The Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Karen S Anderson
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ, 85287-4501, USA. .,Center for Personalized Diagnostics, The Biodesign Institute, Arizona State University, Tempe, AZ, USA.
| | - Melissa A Wilson
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ, 85287-4501, USA. .,Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
12
|
Cha JH, Yang WH, Xia W, Wei Y, Chan LC, Lim SO, Li CW, Kim T, Chang SS, Lee HH, Hsu JL, Wang HL, Kuo CW, Chang WC, Hadad S, Purdie CA, McCoy AM, Cai S, Tu Y, Litton JK, Mittendorf EA, Moulder SL, Symmans WF, Thompson AM, Piwnica-Worms H, Chen CH, Khoo KH, Hung MC. Metformin Promotes Antitumor Immunity via Endoplasmic-Reticulum-Associated Degradation of PD-L1. Mol Cell 2018; 71:606-620.e7. [PMID: 30118680 PMCID: PMC6786495 DOI: 10.1016/j.molcel.2018.07.030] [Citation(s) in RCA: 561] [Impact Index Per Article: 80.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/26/2018] [Accepted: 07/23/2018] [Indexed: 11/17/2022]
Abstract
Metformin has been reported to possess antitumor activity and maintain high cytotoxic T lymphocyte (CTL) immune surveillance. However, the functions and detailed mechanisms of metformin's role in cancer immunity are not fully understood. Here, we show that metformin increases CTL activity by reducing the stability and membrane localization of programmed death ligand-1 (PD-L1). Furthermore, we discover that AMP-activated protein kinase (AMPK) activated by metformin directly phosphorylates S195 of PD-L1. S195 phosphorylation induces abnormal PD-L1 glycosylation, resulting in its ER accumulation and ER-associated protein degradation (ERAD). Consistently, tumor tissues from metformin-treated breast cancer patients exhibit reduced PD-L1 levels with AMPK activation. Blocking the inhibitory signal of PD-L1 by metformin enhances CTL activity against cancer cells. Our findings identify a new regulatory mechanism of PD-L1 expression through the ERAD pathway and suggest that the metformin-CTLA4 blockade combination has the potential to increase the efficacy of immunotherapy.
Collapse
MESH Headings
- AMP-Activated Protein Kinases/genetics
- AMP-Activated Protein Kinases/immunology
- Animals
- Antineoplastic Agents/pharmacology
- B7-H1 Antigen/genetics
- B7-H1 Antigen/immunology
- CTLA-4 Antigen/genetics
- CTLA-4 Antigen/immunology
- Cell Line, Tumor
- Endoplasmic Reticulum/drug effects
- Endoplasmic Reticulum/genetics
- Endoplasmic Reticulum/metabolism
- Endoplasmic Reticulum-Associated Degradation
- Epithelial Cells/cytology
- Epithelial Cells/drug effects
- Epithelial Cells/immunology
- Female
- Gene Expression Regulation, Neoplastic
- Glycosylation
- Humans
- Hypoglycemic Agents/pharmacology
- Mammary Glands, Human/cytology
- Mammary Glands, Human/drug effects
- Mammary Glands, Human/immunology
- Melanoma, Experimental/drug therapy
- Melanoma, Experimental/genetics
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Metformin/pharmacology
- Mice
- Mice, Inbred NOD
- Phosphorylation
- Serine/metabolism
- T-Lymphocytes, Cytotoxic/cytology
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
Collapse
Affiliation(s)
- Jong-Ho Cha
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul 151-742, Korea
| | - Wen-Hao Yang
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Weiya Xia
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yongkun Wei
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Li-Chuan Chan
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Graduate School of Biomedical Sciences, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Seung-Oe Lim
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chia-Wei Li
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Taewan Kim
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shih-Shin Chang
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Heng-Huan Lee
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jennifer L Hsu
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, China Medical University, Taichung 404, Taiwan; Department of Biotechnology, Asia University, Taichung 413, Taiwan
| | - Hung-Ling Wang
- Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, China Medical University, Taichung 404, Taiwan
| | - Chu-Wei Kuo
- Institute of Biological Chemistry, Academia Sinica, Nankang, Taipei 115, Taiwan
| | - Wei-Chao Chang
- Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, China Medical University, Taichung 404, Taiwan; Genomics Research Center, Academia Sinica, Nankang, Taipei 115, Taiwan
| | - Sirwan Hadad
- Department of Surgery, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Colin A Purdie
- Department of Pathology, Ninewells Hospital and Medical School, Dundee DD1 9SY, UK
| | - Aaron M McCoy
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shirong Cai
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yizheng Tu
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jennifer K Litton
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Elizabeth A Mittendorf
- Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Stacy L Moulder
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - William F Symmans
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Alastair M Thompson
- Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Helen Piwnica-Worms
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Graduate School of Biomedical Sciences, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Chung-Hsuan Chen
- Genomics Research Center, Academia Sinica, Nankang, Taipei 115, Taiwan
| | - Kay-Hooi Khoo
- Institute of Biological Chemistry, Academia Sinica, Nankang, Taipei 115, Taiwan
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Graduate School of Biomedical Sciences, University of Texas Health Science Center, Houston, TX 77030, USA; Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, China Medical University, Taichung 404, Taiwan; Department of Biotechnology, Asia University, Taichung 413, Taiwan.
| |
Collapse
|
13
|
Liu Q, Das M, Liu Y, Huang L. Targeted drug delivery to melanoma. Adv Drug Deliv Rev 2018; 127:208-221. [PMID: 28939379 DOI: 10.1016/j.addr.2017.09.016] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 08/29/2017] [Accepted: 09/14/2017] [Indexed: 12/21/2022]
Abstract
Melanoma derived from melanocytes is the most aggressive genre of skin cancer. Although the considerable advancement in the study of human cancer biology and drug discovery, most advanced melanoma patients are inevitably unable to be cured. With the emergence of nanotechnology, the use of nano-carriers is widely expected to alter the landscape of melanoma treatment. In this review, we will discuss melanoma biology, current treatment options, mechanisms behind drug resistance, and nano-based solutions for effective anti-cancer therapy, followed by challenges and perspectives in both pre-clinical and clinical settings.
Collapse
Affiliation(s)
- Qi Liu
- Division of Pharmacoengineering and Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; UNC & NCSU Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Manisit Das
- Division of Pharmacoengineering and Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yun Liu
- Division of Pharmacoengineering and Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; UNC & NCSU Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
14
|
Zhao ZB, Long J, Zhao YY, Yang JB, Jiang W, Liu QZ, Yan K, Li L, Wang YC, Lian ZX. Adaptive immune cells are necessary for the enhanced therapeutic effect of sorafenib-loaded nanoparticles. Biomater Sci 2018; 6:893-900. [DOI: 10.1039/c8bm00106e] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Sorafenib is a kinase inhibitor approved for the treatment of primary kidney cancer, advanced primary liver cancer, and radioactive iodine resistant advanced thyroid carcinoma.
Collapse
|
15
|
Liu Q, Zhu H, Liu Y, Musetti S, Huang L. BRAF peptide vaccine facilitates therapy of murine BRAF-mutant melanoma. Cancer Immunol Immunother 2017; 67:299-310. [PMID: 29094184 DOI: 10.1007/s00262-017-2079-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 10/09/2017] [Indexed: 02/07/2023]
Abstract
Approximately, 50% of human melanomas are driven by BRAF mutations, which produce tumors that are highly immunosuppressive and often resistant to vaccine therapy. We introduced lipid-coated calcium phosphate nanoparticles (LCP NPs) as a carrier to efficiently deliver a tumor-specific antigen, the BRAFV600E peptide, to drive dendritic cell (DC) maturation and antigen presentation in C57BL6 mice. The BRAF peptide vaccine elicited a robust, antigen-specific cytotoxic T cell response and potent tumor growth inhibition in a murine BRAF-mutant melanoma model. Advanced BRAF-specific immune response was illustrated by IFN-γ production assay and cytotoxic T lymphocyte (CTL) assay. Remodeling of immunosuppressive modules within the tumor microenvironment further facilitated CTL infiltration. Thus, using LCP NPs to deliver the BRAF peptide vaccine is a promising strategy for the BRAF-mutant melanoma therapy.
Collapse
Affiliation(s)
- Qi Liu
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 1315 Kerr Hall, Campus Box 7571, Chapel Hill, NC, 27599, USA
| | - Hongda Zhu
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 1315 Kerr Hall, Campus Box 7571, Chapel Hill, NC, 27599, USA.,School of Food and Biology Engineering, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China
| | - Yun Liu
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 1315 Kerr Hall, Campus Box 7571, Chapel Hill, NC, 27599, USA
| | - Sara Musetti
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 1315 Kerr Hall, Campus Box 7571, Chapel Hill, NC, 27599, USA
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 1315 Kerr Hall, Campus Box 7571, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
16
|
Naqash AR, Stroud G, Collichio FA, Muzaffar M, Sharma N, Walker P. Metastatic melanoma in a 95 years old patient responding to treatment with talimogene laherparepvec followed by nivolumab. Acta Oncol 2017; 56:1327-1330. [PMID: 28481677 DOI: 10.1080/0284186x.2017.1324212] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Abdul R. Naqash
- Division of Hematology/Oncology, East Carolina University, Greenville, NC, USA
| | - Geoffrey Stroud
- Division of Hematology/Oncology, East Carolina University, Greenville, NC, USA
| | - Frances A. Collichio
- Division of Hematology/Oncology, Lineberger Comprehensive Center, University of North Carolina, NC, USA
| | - Mahvish Muzaffar
- Division of Hematology/Oncology, East Carolina University, Greenville, NC, USA
| | - Nitika Sharma
- Division of Hematology/Oncology, East Carolina University, Greenville, NC, USA
| | - Paul Walker
- Division of Hematology/Oncology, East Carolina University, Greenville, NC, USA
| |
Collapse
|
17
|
Complete response to anti-PD-1 nivolumab in massive skin metastasis from melanoma. Anticancer Drugs 2017; 28:808-810. [DOI: 10.1097/cad.0000000000000515] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
18
|
Rouas-Freiss N, LeMaoult J, Verine J, Tronik-Le Roux D, Culine S, Hennequin C, Desgrandchamps F, Carosella ED. Intratumor heterogeneity of immune checkpoints in primary renal cell cancer: Focus on HLA-G/ILT2/ILT4. Oncoimmunology 2017; 6:e1342023. [PMID: 28932645 DOI: 10.1080/2162402x.2017.1342023] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 05/22/2017] [Accepted: 06/07/2017] [Indexed: 02/08/2023] Open
Abstract
The establishment and maintenance of anti-tumor immune responses are the objectives of cancer immunotherapy. Despite recent promising advances, the effectiveness of these approaches has been limited by the multiple immunosuppressive mechanisms developed by tumors (checkpoint). The aim of the present study was to demonstrate intratumor heterogeneity at the levels of immune escape strategies and tumor-host relationships. We focused on well-known checkpoints such as PD1/PDL1 and on a new checkpoint involving HLA-G and its receptors ILT2/ILT4. A prospective study was performed on 19 renal-cell carcinoma patients that were included during hospitalization for surgical tumor resection. Different areas of the tumor were collected for each patient and subjected to both immunohistochemical and flow cytometry analysis. Immune cells from peripheral blood were concomitantly analyzed for each patient. Our results show the heterogeneous expression of PD1/PDL1 and HLA-G/ILT in the various areas of the same tumor. Intratumor heterogeneity was found both at tumor cell and infiltrating immune cell levels. From a clinical point of view, this work highlights the functional redundancies of checkpoints and the need to adapt personalized poly-immunotherapy.
Collapse
Affiliation(s)
- Nathalie Rouas-Freiss
- CEA, Direction de la Recherche Fondamentale (DRF), Service de Recherche en Hemato-Immunologie (SRHI), Paris, France.,Universite Paris Diderot, Sorbonne Paris Cite, IUH, Hopital Saint-Louis, UMR_E5, Paris, France
| | - Joel LeMaoult
- CEA, Direction de la Recherche Fondamentale (DRF), Service de Recherche en Hemato-Immunologie (SRHI), Paris, France.,Universite Paris Diderot, Sorbonne Paris Cite, IUH, Hopital Saint-Louis, UMR_E5, Paris, France
| | - Jérôme Verine
- CEA, Direction de la Recherche Fondamentale (DRF), Service de Recherche en Hemato-Immunologie (SRHI), Paris, France.,Universite Paris Diderot, Sorbonne Paris Cite, IUH, Hopital Saint-Louis, UMR_E5, Paris, France.,Service d'Anatomo-Pathologie, AP-HP, Hopital Saint-Louis, Paris, France
| | - Diana Tronik-Le Roux
- CEA, Direction de la Recherche Fondamentale (DRF), Service de Recherche en Hemato-Immunologie (SRHI), Paris, France.,Universite Paris Diderot, Sorbonne Paris Cite, IUH, Hopital Saint-Louis, UMR_E5, Paris, France
| | - Stéphane Culine
- CEA, Direction de la Recherche Fondamentale (DRF), Service de Recherche en Hemato-Immunologie (SRHI), Paris, France.,Universite Paris Diderot, Sorbonne Paris Cite, IUH, Hopital Saint-Louis, UMR_E5, Paris, France.,Service d'Oncologie Médicale, AP-HP, Hopital Saint-Louis, Paris, France
| | - Christophe Hennequin
- CEA, Direction de la Recherche Fondamentale (DRF), Service de Recherche en Hemato-Immunologie (SRHI), Paris, France.,Universite Paris Diderot, Sorbonne Paris Cite, IUH, Hopital Saint-Louis, UMR_E5, Paris, France.,Service de Radiothérapie, AP-HP, Hopital Saint-Louis, Paris, France
| | - François Desgrandchamps
- CEA, Direction de la Recherche Fondamentale (DRF), Service de Recherche en Hemato-Immunologie (SRHI), Paris, France.,Universite Paris Diderot, Sorbonne Paris Cite, IUH, Hopital Saint-Louis, UMR_E5, Paris, France.,Service d'Urologie, AP-HP, Hopital Saint-Louis, Paris, France
| | - Edgardo D Carosella
- CEA, Direction de la Recherche Fondamentale (DRF), Service de Recherche en Hemato-Immunologie (SRHI), Paris, France.,Universite Paris Diderot, Sorbonne Paris Cite, IUH, Hopital Saint-Louis, UMR_E5, Paris, France
| |
Collapse
|
19
|
Huang J, Liu F, Liu Z, Tang H, Wu H, Gong Q, Chen J. Immune Checkpoint in Glioblastoma: Promising and Challenging. Front Pharmacol 2017; 8:242. [PMID: 28536525 PMCID: PMC5422441 DOI: 10.3389/fphar.2017.00242] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 04/18/2017] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma (GBM) is a severe malignant brain cancer with poor overall survival. Conventional intervention remains dismal to prevent recurrence and deterioration of GBM cell. Recent years have witnessed exciting breakthroughs in novel immune strategies, especially checkpoint inhibitors, some of which have become adjuvant setting after standard of care in melanoma. Several clinical trials of checkpoint inhibitors are ongoing in glioblastoma and other brain carcinomas. Plus, synergistic combinations of checkpoint inhibitors with conventional therapy strategies—radiotherapy, temozolomide, bevacizumab, and corticosteroids are now being exploited and applied in clinical settings. This review highlights the recent developments of checkpoints in GBM immunotherapy to provide a brief and comprehensive review of current treatment options. Furthermore, we will discuss challenges remained, such as unique immune system of central nervous system (CNS), immune-related toxicities, synergies, and adverse interactions of combination therapies.
Collapse
Affiliation(s)
- Jing Huang
- Department of Psychiatry, the Second Xiangya Hospital, Central South UniversityChangsha, China.,Mental Health Institute of the Second Xiangya Hospital, Chinese National Clinical Research Center on Mental Disorders (Xiangya), Chinese National Technology Institute on Mental Disorders, Hunan Key Laboratory of Psychiatry and Mental Health, Central South UniversityChangsha, China
| | - Fangkun Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University (CSU)Changsha, China
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University (CSU)Changsha, China
| | - Hui Tang
- Department of Psychiatry, the Second Xiangya Hospital, Central South UniversityChangsha, China.,Mental Health Institute of the Second Xiangya Hospital, Chinese National Clinical Research Center on Mental Disorders (Xiangya), Chinese National Technology Institute on Mental Disorders, Hunan Key Laboratory of Psychiatry and Mental Health, Central South UniversityChangsha, China
| | - Haishan Wu
- Department of Psychiatry, the Second Xiangya Hospital, Central South UniversityChangsha, China.,Mental Health Institute of the Second Xiangya Hospital, Chinese National Clinical Research Center on Mental Disorders (Xiangya), Chinese National Technology Institute on Mental Disorders, Hunan Key Laboratory of Psychiatry and Mental Health, Central South UniversityChangsha, China
| | - Qianni Gong
- Department of Minimally Invasive Surgery, the Second Xiangya Hospital, Central South UniversityChangsha, China
| | - Jindong Chen
- Department of Psychiatry, the Second Xiangya Hospital, Central South UniversityChangsha, China.,Mental Health Institute of the Second Xiangya Hospital, Chinese National Clinical Research Center on Mental Disorders (Xiangya), Chinese National Technology Institute on Mental Disorders, Hunan Key Laboratory of Psychiatry and Mental Health, Central South UniversityChangsha, China
| |
Collapse
|